Next Article in Journal
Enlargement of the Nerve Fibers of Silenced Lumbosacral Motoneurons in Cats
Previous Article in Journal
Non Coding RNAs as Regulators of Wnt/β-Catenin and Hippo Pathways in Arrhythmogenic Cardiomyopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical and Research MRI Techniques for Assessing Spinal Cord Integrity in Degenerative Cervical Myelopathy—A Scoping Review

1
Spine Labs, St. George & Sutherland Clinical School, UNSW Faculty of Medicine, Kogarah, NSW 2217, Australia
2
Faculty of Medicine, University of New South Wales, Kensington, NSW 2052, Australia
3
Spine Service, Department of Orthopaedic Surgery, St. George Hospital Campus, Kogarah, NSW 2217, Australia
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(10), 2621; https://doi.org/10.3390/biomedicines10102621
Submission received: 25 August 2022 / Revised: 5 October 2022 / Accepted: 11 October 2022 / Published: 18 October 2022
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Background: Degenerative cervical myelopathy (DCM) manifests as the primary cause of spinal cord dysfunction and is non-traumatic, chronic and progressive in nature. Decompressive surgery is typically utilised to halt further disability and neurological dysfunction. The limitations of current diagnostic options surrounding assessment and prognostic potential render DCM still largely a clinical diagnosis. Aims: To outline the limitations of current diagnostic techniques, present evidence behind novel quantitative MRI (qMRI) techniques for assessing spinal cord integrity in DCM and suggest future directions. Method: Articles published up to November 2021 were retrieved from Medline, EMBASE and EBM using key search terms: spinal cord, spine, neck, MRI, magnetic resonance imaging, qMRI, T1, T2, T2*, R2*, DTI, diffusion tensor imaging, MT, magnetisation transfer, SWI, susceptibility weighted imaging, BOLD, blood oxygen level dependent, fMRI, functional magnetic resonance imaging, functional MRI, MRS, magnetic resonance spectroscopy. Results: A total of 2057 articles were retrieved with 68 articles included for analysis. The search yielded 2 articles on Quantitative T1 mapping which suggested higher T1 values in spinal cord of moderate-severe DCM; 43 articles on DTI which indicated a strong correlation of fractional anisotropy and modified Japanese Orthopaedic Association scores; 15 articles on fMRI (BOLD) which demonstrated positive correlation of functional connectivity and volume of activation of various connections in the brain with post-surgical recovery; 6 articles on MRS which suggested that Choline/N-acetylaspartate (Cho/NAA) ratio presents the best correlation with DCM severity; and 4 articles on MT which revealed a preliminary negative correlation of magnetisation transfer ratio with DCM severity. Notably, most studies were of low sample size with short timeframes within 6 months. Conclusions: Further longitudinal studies with higher sample sizes and longer time horizons are necessary to determine the full prognostic capacity of qMRI in DCM.

1. Introduction

1.1. Epidemiology

Degenerative cervical myelopathy (DCM), earlier referred to as cervical spondylotic myelopathy (CSM), is the most common cause of spinal cord dysfunction, affecting an estimated 5% of adults over 40 years of age [1]. It is a significant cause of disability and carries substantial economic burden to the affected individuals, including their families and their community [2].
A comprehensive review of the literature demonstrated that such degenerative conditions of the spine are estimated to encompass 59% of non-traumatic spinal cord injury in Japan [3], 54% in the United States [4], 18–26% in Australia [5,6,7] and 16–39% in Europe [8,9,10,11,12,13,14]. The incidence was also purported to be 76, 26 and 6 per million in North America, Europe and Australia, respectively [7]. Notably, these data exclude many patients with less severe symptoms. Very few studies have been conducted on DCM prevalence. In Victoria, Australia, the prevalence of all non-traumatic spinal cord injury was estimated to be 367.2 per million in 2010 [15]. On the basis of these figures, studies have estimated the incidence and prevalence of DCM in the United States to be 41 and 605 per million, respectively [16].
However, a constraining factor of these estimates are the poor quality of the data of which they are derived from, and it is likely that the figures severely underestimate the burden of disease. As one of the most common causes of non-traumatic spinal-cord injury it is reasonable to infer that DCM represents a formidable issue in the aging population [17].

1.2. Natural History

AO Spine RECODE-DCM has recently listed the diagnosis and pathogenesis, as some of the top priorities in DCM research [18]. Degenerative cervical myelopathy is a degenerative condition and as such, it is non-traumatic, chronic and progressive in nature, with surgery traditionally utilised to halt further disability and neurological dysfunction [19]. The pathogenesis of DCM is purported to involve a myriad of static and dynamic factors (Figure 1). Static factors include spinal canal compression, spine deformity, disc herniation, osteophyte formation, ossification of the posterior longitudinal ligaments (OPLL) and ossification of the ligamentum flavum (OFL). Owing to its mobility, the vertebral column of the neck also suffers from dynamic stressors that include biomechanical changes, invagination of the ligamentum flavum and microstructural mechanical spinal cord damage from cervical instability. Such stressors, at a chronic magnitude, induce direct neuronal and glial cell damage as well as a secondary ischaemic cascade of neuronal excitotoxicity and apoptosis which contribute to the development of DCM [17].
Notably, very little is currently known with regards to the exact molecular mechanisms underlying the process of this condition. A 1963 retrospective study of the natural history of DCM found that a majority of patients had poor prognosis, 87% of which progressing to moderate or severe disability at the last follow up. Several historical and present day studies have indicated that the disease course of DCM is highly variable [20,21,22,23,24,25,26,27,28,29,30,31,32,33]. In particular, a 1956 study of 120 DCM patients and found 5% patients had a rapid onset of symptoms followed by long periods of quiescence, 20% had a slow, progressive deleteriousness of neurofunction, and 75% had a stepwise decline of neurofunction [21]. A further 2017 systematic review demonstrated that 20–67% of patients had experienced neurological deterioration after 3–6 years of follow-up [34]. It is not yet clear what manifests these differences in DCM pathogenesis between patients. Further research into DCM at a molecular level could result in promising diagnostic methods, enable detection at early stages and thus render timely intervention and treatment.
The current data associated with the natural history of DCM is largely derived from low-quality retrospective studies [19,35]. The limited existing prospective studies are markedly underpowered and have low level evidence with inconsistent results and risks of bias [36]. Accordingly, there persists a necessity for a large-scale prospective study focusing on natural history of DCM, specifically with the inclusion of novel multiparametric quantitative MRI that will be discussed further on.

1.3. Current Diagnostic Options and Limitations

1.3.1. Clinical

Diagnosis of DCM typically necessitates a congruity between clinical (Table 1.) and investigatory findings (Figure 2). A thorough history and physical examination should first ensue when DCM is suspected.
Limitations: Although a useful element of diagnosis, physical tests are not always consistent in their ability to quantify the severity of DCM which is an important element in the consideration of surgical intervention [44]. Thus, correlations in further investigatory measures are required to arrive at the correct diagnosis.

1.3.2. Scoring Systems

Clinicians utilise scoring systems to categories the functional impairment of various conditions. Whilst different classifications may arise, there typically exists one standardised system for publications and treatments. This is not true for DCM; whereby different systems are utilised based upon preference. A 2016 systematic-review revealed that reported outcomes varied widely between studies of DCM [45]. Table 2 details the current most common classification systems, their benefits and limitations. The mJOA scale followed by the Nurick Grading system are the current most widely adopted measure for DCM patients [46].
However, the limitations covered in Table 2, are particularly problematic in mild DCM whereby strong floor and ceiling effects[e] in these scales inhibit ascertainment of more subtle neurological changes that provide information for decision-making in surgery18. As such, there exists the need to develop both a standardised scoring system and more sensitive and objective outcome instruments to enable more effective clinical assessment and efficient synthesis of research.

1.3.3. Conventional MRI

Conventional MRI is the primary modality utilised for imaging in DCM as it enables high-resolution depiction of neural structures, bone and ligaments that are difficult to visualise in other scans [57]. Conventional MRI (such as T1-weighted and T2-weighted imaging) can characterise the degree and nature of degeneration (i.e., OPLL, spondylosis, disc herniation, hypertrophy of ligamentum flavum), identify spinal-cord compression, highlight changes in spinal-canal diameter, and detect changes in signal intensity [58,59,60]. MRI can also assist in ruling-out resembling differentials or other causes of myelopathy such as a tumour syringomyelia or demyelinating plaques [2,38,42]. CT myelography should be utilised in situations of MRI contraindication [61].
Identifying spinal-cord compression plays a pivotal role in treatment selection and outcome prediction and thus should be the foremost investigation. It is typically described based on the number of compression sites [30], appearance [32,62,63,64,65] or ratio between the anteroposterior diameter and the transverse diameter (CR = Compression Ratio) [66,67]. A maximum spinal-cord compression (MSCC) index has also been developed by Fehlings et al. as a measurement of spinal-cord compression [68]. The primary object of these methods is to determine severity of spinal-cord compression.
Measurements of the anterior-posterior diameter at the region of interest (ROI) can be undertaken to evaluate the degree of spinal-stenosis [30]. Similar to MSCC, Fehlings et al. have developed a protocol to assess the maximum canal compromise (MCC) post-traumatic cervical spine-injury [69]. This has been additionally utilised for degenerative conditions and functions by calculating the canal size at the ROI and analysing it in conjunction to the average canal size for levels above and below. Multi-level signal-intensity changes are suggestive of necrosis or cavitation in the spinal cord and lend to poorer surgical outcomes [70,71,72,73]. T2-hyperintensity in conjunction with T1-hypointensity is associated with greater clinical deterioration when compared to T2-hyperintensity alone due to signal changes in T1-weighted images indicative of more permanent insult [70,74,75,76,77].
Limitations: Findings on conventional MRI do not typically correlate well with the variable clinical presentations of DCM [42]. Although spinal-cord compression is a sensitive marker of myelopathy [78], approximately 5% of asymptomatic patients also present with it [42], thereby limiting its specificity. The supine patient positioning in conventional MRI hinders its utility in assessing alignment, providing only a superficial assessment for situations in which upright films are not available [79]. Conventional MRI is intrinsically limited in its capability to characterize tissue injury in the spinal-cord because of the lack of specificity in T1/T2WI signal-change and cannot highlight specific pathophysiological processes at a cellular level (demyelination, axonal loss, inflammation, oedema, gliosis and apoptosis) [57]. It also is not a good predictor of neurologic function before/after surgical intervention and has low sensitivity for structural spinal cord change in cervical myelopathy [57,70,80,81,82,83].

1.3.4. Plain Radiographs and Computed Tomography (CT)

Computed Tomography is useful for the study of bone anatomy and can aid in cases where spinal-fusion is being considered as a treatment. In cases where MRI is contraindicated (such as the presence of pacemakers or other internal metallic objects), CT is a valuable imaging alternative. Plain radiographs can provide useful information about spinal-canal stenosis, degenerating discs, degenerating joints, OPLL, vertebrae fusion, cervical-spine alignment and subluxation [2,38,84,85]. This can reveal scoliosis and loss of physiological cervical-lordosis and kyphosis. Lateral-films in cervical-flexion and extensions are utilised to evaluate instability of the cervical-spine. DCM patients frequently showcase increased C2-C7 Cobb angles, upper T1 slopes, lower C7 slopes and upper C7 slopes [86].
Limitations: Computed tomography suffers the same inability to characterise tissue injury that conventional MRI does [57]. In addition, a 2017 systematic-review found that the overall strength of evidence regarding the predictive value that CT parameters have for the clinical presentation or outcome of DCM is low [87]. There is also the issue of radiation exposure. Overall CT and plain radiographs play a more complementary role in DCM diagnosis, acting as an alternative to MRI and aiding in surgical-planning [88].

1.3.5. Electrophysiology

Several studies have indicated good correlation between electrophysiology and the severity of myelopathy, presenting it as a reliable predictor of surgical-outcomes [89]. Somatosensory evoked-potentials (SEPs) and motor evoked-potentials (MEPs) can be, respectively, utilised to detect central sensory conduction impairment and prolonged motor latency in DCM [2,89,90]. They are also useful in detecting subclinical degenerative spinal-cord compression in asymptomatic patients and are thus useful in early identification of patients likely to develop myelopathy [91,92,93,94]. Feng et al. reported a correlation between the SEP and a declining mJOA (a more severe deficit) in an investigation of progressive myelopathy [95]. Needle electromyography (EMG) is a highly sensitive indicator of anterior horn cells damage, which occurs due to compression and ischemia in DCM [96]. Nerve-conduction studies can also be used to rule out peripheral neuropathy and nerve-entrapment [2]. These techniques also allow other neuromuscular diseases that can mimic DCM to be ruled out (motor neurone disease, ALS) [97,98]. Apart from aiding in diagnosis and preoperative evaluation, electrophysiology facilitates longitudinal assessment. Capone et al. found that a decrease in central-motor conduction time for the tibialis-anterior muscle correlated with an increased mJOA score post-surgery. It therefore concluded that the beneficial effects of spinal-cord surgery could be detected with MEP, making it a useful tool in determining efficacy of post-operative rehabilitation [99].
Limitations: Electrophysiology provides no anatomical information and thus cannot determine the exact location of the lesion [100]. Although some evidence exists to justify the effectiveness of electrophysiology in predicting operative outcomes, the area remains to be better defined. A systematic review found a decrease in electrophysiology publications compared with other domains of DCM, suggesting a declining interest in this area [101]. Additional studies would be required before it can be universally recommended.

1.4. Novel qMRI Modalities and Parameters

The limitations of current diagnostic options render DCM still largely a clinical diagnosis [17], making it necessary to develop and further research on novel diagnostic options with objective quantitative measures.
Advanced novel MRI protocols have been developed for the spinal cord that allow for acquisition within 45 min [102]. This involves direct measurement of spinal cord tissue changes, demyelination, axonal-injury and atrophy and thus renders the attainment of quantitative microstructural sequences now possible in the context of DCM. Such qMRI sequences and their derivable quantitative metrics are highlighted in Table 3. These derived metrics are highly sensitive to the myelopathic progression and can allow for the realisation of subclinical tissue-damage in patients with asymptomatic cervical-cord compression [103,104,105,106,107]. Quantitative metrics derived from DWI, such as DTT and DTI have been found to be more valuable when compared to conventional MRI scans in aiding diagnosis and outcome prediction in patients with DCM [108,109]. These qMRI sequences will be expanded upon later in this review. As an emerging field, the development of more advanced imaging techniques may potentiate in superior diagnostic tools, improved correlation with impairment and long-term predictions of DCM outcomes.

1.5. Objective

A scoping review was conducted in order to systematically map the research done in this area, as well as to identify any existing gaps in knowledge. The following research question was formulated:
‘What is known from the literature about existing clinical and novel research MRI techniques for assessing spinal cord integrity in patients with Degenerative Cervical Myelopathy (DCM)?’

2. Methodology

2.1. Data Sources

Articles published up to November 2021 were retrieved from three main databases: Medline, EMBASE and EBM. Combinations and variations of keywords were used to conduct a comprehensive search: spinal cord, spine, neck, magnetic resonance imaging, MRI, qMRI, T1, T2, T2*, R2*, DTI, diffusion tensor imaging, MT, magnetisation transfer, SWI, susceptibility weighted imaging BOLD, blood oxygen level dependent, fMRI, functional MRI, functional magnetic resonance imaging, MRS, magnetic resonance spectroscopy.

2.2. Selection Criteria

Papers to be included required a focus on quantitative MRI techniques for assessing spinal cord integrity in patients with DCM. Exclusion criteria included any articles focusing on non-myelopathic diseases, flexion induced myelopathy, conventional MRI techniques only, non-degenerative causes of myelopathy, and studies on asymptomatic patients only. Reviews and non-English articles were also excluded from this study. A detailed flow diagram of this method can be seen in Figure 3.

2.3. Synthesis of Results

Following the process detailed in Figure 3., after studies were included for qualitative review detailed were collected in a tabular format (see Appendix CTable A8). Details were then summarised (see Table 4 in Section 3) and underwent discussion and critical appraisal in Section 4.

3. Results

A total of 2055 articles were identified using the search strategy outlined in Figure 3 from the three databases. There were 283 duplicates removed by automation and 99 removed manually. The remaining 1770 records were then screened by title and abstract according to the inclusion and exclusion criteria leaving 155 articles to be screened for full-text eligibility. A detailed evaluation rendered 68 articles included for analysis. The complete database search strategy can be found in Appendix B (Figure A1).

4. Discussion

4.1. Quantitative T1 and T2 Mapping

4.1.1. Principles

Quantitative T1/T2 mapping calculates the T1 (spin-lattice or longitudinal relaxation) time or T2 (spin-spin or transverse relaxation) time of a certain tissue and displays them on a parametric map. This is in contrast to conventional T1/T2-weighted MRI, which displays differences in T1/T2 relaxation times of tissues as a hyper/hypointense image with limited quantitative output [186,187]. T1 is influenced by the tissue’s water, lipid, protein and iron content which explains its sensitivity to pathological microstructural changes in the spinal cord [125,186]. T2 is influenced primarily by the water content of the tissue and thus increases in T2 is associated with increased water content pertaining to increased disc water or glycosaminoglycan content in the spinal-cord [187].

4.1.2. Application in DCM

Despite being a fundamental parameter in MR imaging, T1/T2 mapping is not traditionally frequented in clinical practice due to its lengthy scan times. However, recent advances to speed of acquisition have enabled T1/T2 to become another viable option in the analysis of the pathological spinal cord [120,188,189,190,191]. Notably, T1 has been utilised in a multitude of brain studies to investigate changes in white and grey matter ratio in both a physiological and pathological context of which has demonstrated an interesting clinical correlation with cognitive decline [192,193]. Much of the literature surrounding its use in the spinal cord have highlighted reasonable scan times and reproducible methods to measure the T1 values in the healthy cervical spinal cord [120,194]. T2 relaxation times have been shown to correlate with lumbar disc degeneration [195,196,197], however our literature search reveals no studies conducted on the cervical spine. It thus pertains that T2 may potentiate as a useful biomarker for analysing spinal-cord integrity in DCM and so future research combining the two may prove beneficial.
From the conducted literature search, there exists only two studies applying T1 to the degenerative cervical spinal-cord which utilised 2D single section (inversion-recovery) radial-gradient echo [151] and 3D-MP2RAGE [125] sequences. The former study demonstrated that the difference in T1 relaxation times between stenotic segments and non-stenotic segments above and below were longer in higher grades of stenosis. It found that absolute T1 values were higher in high-grade stenosis but found inconclusive correlation between lower grades of stenosis [151]. The latter study utilising 3D-MP2RAGE found higher T1 values in the overall spinal-cord and the level of compression of patients with moderate-severe-DCM compared to healthy controls [125]. Overall, correlation findings of T1 and clinical severity are variable and inconclusive, particularly for patients with mild DCM which still maintain as the most difficult group to diagnose. Both studies are limited by their power and cross-sectional design. Further longitudinal studies with higher-sample sizes should be conducted particularly with respect to post-operative outcome to reach more conclusive results.

4.2. Diffusion Tensor Imaging (DTI)

4.2.1. Principles

Diffusion tensor imaging is a sensitive measure of tissue microstructure that works by measuring the diffusion of water molecules. Water in a glass of water for example would be considered isotropic, the diffusion would be the same in every direction. The diffusion of water in tissue however would be considered anisotropic, whereby the diffusion will vary with direction. This would depend on the tissue type, integrity, architecture and presence of barriers. Fractional anisotropy (FA) (a quantifiable parameter of DTI) is a value between 0 and 1 that indicates the degree to which the diffusion of water is limited to one axis. Notably in a healthy spinal cord, the axons largely run in in one direction—commonly analogised to a bundle of straws. In DCM, the axonal integrity is compromised and by first principles, fractional anisotropy would be reduced. Other quantifiable DTI parameters include mean diffusivity (MD) or apparent diffusion coefficient which are both measures of the average magnitude of water diffusion within a tissue [102,118].

4.2.2. Application in DCM

Recent studies have indicated a strong correlation between FA (from DTI) and clinical assessments such as mJOA [70,168,198,199,200]. Specifically Dong et al. found that DCM patients presenting with a higher FA score at the level of compression were the most likely group to achieve a better functional recovery after surgical decompression [199]. The conducted literature search indicated consistent findings of the correlation of FA and spinal cord integrity in DCM as well its significant role in prognosis [60,104,107,108,118,124,127,128,131,132,134,135,137,138,139,141,142,143,144,145,146,149,152,153,154,155,157,158,161,162,165,166,167,168,175,176,177,178,179,181,182,183,184,185]. Interestingly, Wang et al. conducted a prospective longitudinal study of 93 DCM patients and 36 healthy-controls and found that DTI parameter ratios (DTI measurement at test cervical level divided the measurement at C1-C2 levels) are more useful than absolute DTI metrics when assessing DCM as absolute metrics can be confounded by age and cervical level [108].
Therefore, DTI can be considered as not only a complementary diagnostic evaluation, but as a vital tool in the diagnosis of DCM and an early identifier of the candidates best suited to surgery. It is important to note however that as an emerging field, many existing studies [198,199,201,202,203] are of low-sample size and could suffer from potential bias from study design limits, patient selection bias or lack of blinding when examining new technologies. Studies utilising 1.5T-MRI as opposed to 3T-MRI are limited by poorer performance and resolution [158,201]. Additionally, some studies [134,158,177,183] are limited by their cross-sectional nature (limiting their ability to predict disease progression) and could benefit from more longitudinal components.
From our conducted literature search, only 3/43 articles were of a prospective longitudinal design with a DCM cohort size of greater than 50. A total of 26/43 of the studies were of a cross-sectional design, and a majority of the longitudinal design studies had a short follow-up period of 3–6 months. As such there exists a need for additional large-scale longitudinal prospective studies to be conducted on DCM utilising DTI methods with longer time horizons and larger sample sizes. DCM is a chronic progressive disease and as it stands there exists no study that correlates longer-term progression (of over 3 years) with DTI parameter scores. Such quantifiable long-term studies could assist in identifying the characteristics of groups at risk of progressing deleteriously, thus contributing to the identification of patients who may benefit from early intervention. Further, in a prospective case control study, that slowed down due to COVID-19, we have standardised protocols (Table 5.) and successfully extracted data from healthy controls (Figure 4). This work will likely shed light on the spectrum of DCM when completed.

4.3. Functional MRI (BOLD)

4.3.1. Principles

Functional-MRI (fMRI) is based on the BOLD contrast mechanism first introduced in 1990 and is a non-invasive technique that allows for the detection of neuronal activity. The fundamental principle behind the BOLD mechanism is that metabolic oxygen demand after neuronal activation causes a rise in blood flow and blood volume. This blood supply surpasses the actual oxygen needs which renders a transient rise of oxyhaemoglobin in the venous compartment and a relative decrease in the concentration of deoxyhaemoglobin. As deoxyhaemoglobin has paramagnetic properties, the change in deoxyhaemoglobin can be measured with MRI [112,204]. In DCM, fMRI has primarily been utilised to measure the functional connectivity (FC) and volume of activation (VOA) of regions of the brain before and after surgery to potentiate correlation [123,126,129,133,136,140,150,160,164,169,172,173,174,180].

4.3.2. Application in DCM

The brain resting-state fMRI has been proven to be successful in differentiating DCM patients from healthy patients [123,126,150,173,174]. In contrast to the aforementioned MRI techniques which measures structural damage/integrity within the conduction pathways, BOLD fMRI measures the functional activation within the brain of which incorporates information collection, interpretation and distribution for all motor and cognitive functions. As a disease with a plethora of motor and sensory symptoms (see Table 1) DCM-associated information would be distributed to widespread areas of the brain [126,173]. As such, a multitude of studies have been performed to determine if fMRI can predict neurological recovery post-decompression surgery. Our literature search revealed that changes in FC strength between different areas of the brain appear to be associated with neurological improvement post-surgery in DCM. Numerous studies found an increase in VOA or FC strength of the pre/postcentral gyrus and SMA (supplementary motor area) following decompression surgery [123,126,133,140,164,174,180]. Functional connectivity alteration between the thalamus and cortex were also demonstrated [160]. Positive correlation of these findings with functional recovery assessed using various DCM grading questionnaires enables fMRI to indirectly assess spinal integrity in DCM patients.
Notably, Takenaka et al. found a positive correlation between post-operative improvement in the 10 s test ([h] The 10 s test: the number of cycles the fingers can repeatedly grip and release in 10 s) and FC of three visual areas and the right superior-frontal gyrus in DCM patients, of which may enable the construction of a predictive formula for recovery potential [173]. Such a connection between visual cortices and DCM was also established in other studies [129,136]. Furthermore, an additional study by Takenaka et al. determined that resting-state amplitude of low-frequency fluctuation could function as a potential prognostic biomarker for DCM [172]. However, their two studies were limited by the use of mass univariate analyses which can only simply measure association. Given the multi-variable nature of fMRI, univariate analyses may miss information associated with DCM pathology and so studies using multivariate patten analysis should be conducted. Moreover, much of the research in the area is preliminary with studies of low sample sizes. Future external validation studies would be necessary for the proposed predictive formulas. None of the prospective studies have looked at the long-term use case of fMRI (over 6 months), thus development is also needed in this regard.

4.4. Magnetic Resonance Spectroscopy (MRS)

4.4.1. Principles

MRS enables the in vivo quantification of metabolite concentration from human tissue. The underlying principle behind MRS is that a proton experiences a slightly distinct magnetic field of which is dependent on its chemical environment. Reliable quantification of metabolites utilising 1.5T MRI scanners have been traditionally limited to N-acetyllaspartate (NAA), choline (Cho) and creatine (Cr). However, recent advances in imaging technology and 3T MRI scanners have enabled measurement of glumatate-glumatine (Glx) and myo-inositols (Ins) [205]. N-acetylaspartate, despite not being a disease-specific marker is a sensitive indicator of axonal integrity due to its ability to be detected early in the disease course. Typically, NAA is expressed as an absolute value or as a ratio with Cho/Cr. The NAA/Cr ratio is generally viewed as a better ratio due to the more constant levels of Cr in the nervous system. However, changes in Cho are also believed to reflect increases in membrane phospholipids due to myelin breakdown from demyelinating diseases [113,118].

4.4.2. Application in DCM

Cross-sectional studies have determined that the Cho/NAA ratio is higher in patients with DCM compared to healthy controls [171] and is significantly correlated with mJOA score [135,163]. As such it provides a potentially clinically useful biomarker for the management of DCM. Ellingson et al. utilised both DTI and MRS data in a combined linear model. The results of this optimised model showcased a higher accuracy in predicting mJOA than DTI and MRS alone [135]. Thus, MRS could find utility in tandem with DTI as a predictive tool. Interestingly, Kowalczyk et al. found that cortical levels of NAA/Cr could also serve as a meaningful biomarker in DCM [147,148]. Nagashima et al. investigated alternative metabolites (lactate, alanine, acetate, glutamate, pyruvate and citrate) and found no significant differences between the myelopathic and control group [156]. The main limitations in the area of MRS is that MR spectroscopic data within the spinal cord is quite difficult to acquire reliably due to patient motion, spinal cord movement (due to the pulsatile flow of CSF) and the difficulties associated with magnetic shimming ([i] Magnetic shimming: the process by which the main magnetic field is made more homogenous) [135,147,148,156,163,171].
Overall, research of MRS application in DCM is quite limited with no new research being conducted in the last six years. Of the articles identified from our literature search, all were cross-sectional and of low-sample size. Further longitudinal work should be done to assess the prognostic potential of MRS in DCM.

4.5. Magnetisation Transfer (MT)

4.5.1. Principles

Magnetisation transfer is a contrast mechanism that relies on the interaction between macromolecule bound hydrogen-protons, namely lipids and lipoproteins, and the free-protons (in free water) normally imaged by MRI. As such MT is able to indirectly probe proteins/lipids. The derived parameter, the magnetisation transfer ratio (MTR) reflects the portion of bound protons. Thus, MTR can be utilised as an indirect marker of demyelination and axonal loss as the MT effect indicates the relative density of protein/lipid macromolecules. This ability to measure myelin and axonal loss in vivo allows for application of MT to demyelinating diseases and degenerative diseases like DCM [115,118].

4.5.2. Application in DCM

MTR has been well-established as a marker of myelin integrity in diseases such as multiple sclerosis [206] and has been shown to correlate with histopathological myelin loss [207]. MT imaging also presents an advantage over diffusion-based imaging in the form of higher signal-to-noise ratio and higher spatial resolution [170]. From our literature search, both Cloney et al. and Suleiman et al. found a negative correlation of MTR with severity of DCM (measured via the mJOA [130] and Nurick score [170], respectively), with pathological patients tending to have a decreased MTR compared with a healthy population. Such could be indicative of DCM associated myelin degradation [130,170]. However, Serbruyns et al. conducted a study that noted a decrease in MTR with aging [208]. The correlation of this with poorer functional tasks suggests that demyelination is associated with age-related decreases in functionality. As DCM is an elderly associated disease, this difficulty of determining causation means that MTR should be primarily interpreted as a quantitative measurement of demyelination from any cause, not just DCM. Paliwal et al. have also determined the prognostic potential of MTR for assessing response to surgery and recovery of DCM patients.
Perhaps the primary shortcoming of the current studies involves the small sample sizes and the limited number of prospective longitudinal studies. Future direction in this area could involve studies of higher sample sizes determining prognostic potential, utilisation of multivariate analysis as opposed to linear correlation, and longer follow up periods to track continued improvement beyond 6 months.

4.6. R2* or 1/T2*—A Promising Biomarker

4.6.1. Principles

R2* MRI measures the ‘observable’ or ‘effective’ T2 (termed T2*) whereby R 2   = 1 T 2 . T2* primarily results from inhomogeneities in the main magnetic field as a result of susceptibility-induced field distortion produced by the tissue placed within the field. In the presence of tissue iron, T2* relaxation time shortens and thus R2* increases (as R 2   = 1 T 2 ). Thus R2* represents a quantifiable measure of tissue iron content, notably via deoxyhaemoglobin, hemosiderin or methemoglobin in tissues and lesions [209,210].

4.6.2. Role of Iron in Neurodegenerative Disorders

Homeostasis of heavy metals, such as iron and calcium are critical for cellular function. Imbalances in levels of iron and calcium have been implicated in various neurological disorders [211]. Iron plays an essential role in physiological functions during the ageing process. It is involved in DNA synthesis and repair, oxygen-transport, mitochondrial respiration, myelin synthesis, neurotransmitter synthesis and metabolism [212]. Abnormalities in homeostasis can induce oxidative damage through generation of reactive oxygen species and result in cellular death [212,213,214].
Past and present studies have indicated the disruption of iron homeostasis in a multitude of neurodegenerative diseases such as multiple sclerosis (MS) [215,216,217,218], Alzhiemer’s disease (AD) [219], Parkinson’s disease (PD) [220], Hallervorden-Spatz syndrome [221] and other pathologies involving iron accumulation in the brain [222]. Additionally, evidence indicates abnormal increases in calcium-signalling in AD [223], PD and amyotrophic lateral sclerosis (ALS) [224].
As a predominant neurodegenerative disorder of the ageing population; these changes in iron/calcium level could be implicated in DCM. As it stands; there is no current research in this area and studies looking to quantify these levels could assist in developing new diagnostic options and aid in understanding of the pathological processes of DCM at a molecular level.

4.6.3. Application in DCM

A review of the literature revealed a plethora of studies that were successful in utilising R2*-MRI to quantify iron levels in brain for conditions such as AD [223,225,226,227], PD [228,229,230,231] and MS [232,233,234,235] in an effort to gauge correlation with the disease. A 2018 study utilised this R2*-MRI to quantify iron accumulation following acute traumatic spinal-cord injury [236] and found an increase in brain and brainstem iron accumulation following progressive neurodegeneration of patients. This study however did not explore iron accumulation in the spinal-cord. A 2013 study did however look into iron accumulation in the spinal-cord of mice following traumatic spinal-cord injury and in chronic stages post-injury, using MRI and histological techniques [237]. They were able to detect these iron deposits at the lesion site with live MRI and confirmation with Prussian-blue stains. There has not yet been a study that has looked into spinal-cord iron accumulation in non-traumatic DCM. As the most common cause of spinal-cord dysfunction, a study conducted in this area would prove to be beneficial in developing a new potential MRI biomarker for use in diagnosis.
In Table 6 and Figure 5 we present preliminary unpublished data of R2* ROI scores of the spinal cord of a healthy recruit. These data were obtained through our R2* MRI standardisation protocol and demonstrates the feasibility of this technique to be utilised in the spinal cord of patients. Further work must now be done with regard to DCM patients.

4.7. Quantitative Susceptibility Weighted Imaging (SWI)/Mapping—Another Promising Biomarker

4.7.1. Underlying Principle

Compounds that have paramagnetic, ferromagnetic and diamagnetic properties all interact with the local magnetic field created by MRI. These compounds distort the local magnetic field and alter the phase of the tissue, which ultimately results in a change in signal. SWI is an MRI sequence that is particularly sensitive to such compounds and is therefore commonly used to detect blood products/haemorrhage and calcium. SWI utilises both the effect on phase and the magnitude, unlike conventional MRI sequences. After acquisition, post-processing involves the application of a high-pass filter that removes background inhomogeneity of the magnetic field and the employment of a phase mask which is used to accentuate the change in signal. This culminates in a susceptibility-weighted image which simultaneously incorporates magnitude and phase information for clinical use [238,239,240,241].

4.7.2. Role of Calcium in Neurodegenerative Disorders

Calcium also plays an essential role in the ageing process. Physiological Ca2+ fluxes across plasma membranes and between intracellular compartments play vital roles in neuronal function such as in synaptic-transmission and plasticity, regulating neurite-growth and synaptogenesis, and cell survival. In neurodegenerative disorders these systems are compromised resulting in neuronal degeneration and dysfunction [242,243,244].
Interestingly, studies revealed the role of cellular iron in the stimulation of calcium signalling [245,246,247]. Whilst physiologically, this relationship assists in enhancing calcium-dependent signalling-pathways, an excessive iron accumulation promotes oxidative stress and a pathological upsurge in calcium-signals, of which results in mitochondrial damage. Moreover, this mitochondrial dysfunction renders a loss of iron homeostasis. If uncontrolled, this manifests a deleterious self-perpetuating cycle which eventuates in neuronal death.

4.7.3. Application in DCM

The important self-inducive relationship between iron and calcium renders calcium an important area of research in the scope of neurodegenerative disorders such as DCM. Multiple studies have indicated calcium overload (calcification) at the impact site of acute traumatic spinal-cord injuries [248,249,250]. It has been yet to be determined whether calcium accumulation occurs during the course of DCM. Modern SWI is a MRI sequence that is particularly sensitive to compounds that distort local magnetic-fields and has been successfully utilised to measure calcium accumulation in the brain in vivo [238,251,252,253,254,255]. Of these studies includes a 2010 prospective study [255] with high-sample sizes and a varied population (age and gender). Extending protocols to image the spinal-cord could also prove beneficial in biomarker development.

5. Conclusions and Future Directions

For qMRI to attain clinical significance in DCM it must satisfy three overarching pillars of improvement. Firstly, the necessary advances must be made to minimise issues associated with artifacts and distortions whilst simultaneously improving on image quality, signal-noise ratio and spatial resolution. Such improvements will render qMRI both accurate and able to obtain repeatable results. Secondly, such advances must be utilised to further the literature on DCM, taking in account the limitations of current studies and inadequate areas of research as pointed out in this review. Finally, being both a novel and complex area of study, education is a priority, whereby researchers and clinicians must be updated on these novel quantitative techniques to enable more widespread and effective usage. This in turn will garner further research into this area. Notably, further longitudinal studies with higher sample sizes and longer time horizons are necessary to determine the full prognostic capacity of qMRI in DCM.

Author Contributions

Conceptualisation, B.H., K.S., A.D. (Abhirup Das) and A.D. (Ashish Diwan); search strategy design, B.H. and K.S.; data extraction, B.H.; data validation, K.S.; writing—original draft preparation, B.H.; writing—review and editing, B.H., K.S., A.D. (Abhirup Das) and A.D. (Ashish Diwan). All authors have read and agreed to the published version of the manuscript.

Funding

K.S. is supported by an Australian Government Research Training Program Scholarship, Spine Labs is supported by unrestricted research and education grants by Nuvasive Australia and Baxter Australia, A.Das is supported by internal funds from Spine Service.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We wish to thank the staff at Research Imaging NSW, a core facility located at UNSW for their contribution in preliminary data acquisition (Table 5 and Figure 4).

Conflicts of Interest

No conflict of interest to declare. None of the funding source have a direct or indirect pecuniary interest in the current work.

Abbreviations

1.5TMRI1.5 Tesla magnetic resonance imaging
3TMRI3 Tesla magnetic resonance imaging
ADAlzheimer’s disease
ADCApparent diffusion coefficient
ALSAmyotrophic lateral sclerosis
BOLDBlood oxygen level dependent
ChoCholine
CMSCervical myelopathy scale
CRCompression Ratio
CrCreatine
CSMCervical spondylotic myelopathy
CTComputed tomography
DBSIDiffusion basis spectrum imaging
DCMDegenerative cervical myelopathy
DNADeoxyribonucleic acid
DTIDiffusion tensor imaging
DTTDiffusion tensor tractography
DWIDiffusion weighted imaging
EMGElectromyography
EMSEuropean myelopathy scale
FAFractional anisotropy
FCFunctional connectivity
fFOVFull field of view
fMRIFunctional MRI
GlxGlutamate-glutamine
InsMyo-inositols
MCCMaximum canal compromise
MEPsMotor evoked potentials
mJOAModified Japanese Orthopaedic Association scale
MRIMagnetic resonance imaging
MRSMagnetic resonance spectroscopy
MSMultiple sclerosis
MSCCMaximum spinal cord compression
MTMagnetization transfer
MTRMagnetization transfer ratio
MWFMyelin water fraction
NAAN-acetylaspartate
NCSNerve conduction studies
NDINeck disability index
NPRSNumeric pain rating scale
OPLLOssification of the posterior longitudinal ligaments
PDParkinson’s disease
qMRIQuantitative magnetic resonance imaging
QSMQuantitative susceptibility mapping
R2*MRIR2* magnetic resonance imaging
rFOVReduced field of view
ROIRegion of interest
SMASupplementary motor area
SSEPsSomatosensory evoked potentials
SWISusceptibility weighted imaging
T1WIT1 weighted imaging
T2*WIT2*-weighted imaging
T2WIT2 weighted imaging
VOAVolume of activation

Appendix A. Classification Systems for DCM

Table A1. Modified Japanese Orthopaedic Association (mJOA) Score [256]).
Table A1. Modified Japanese Orthopaedic Association (mJOA) Score [256]).
Modified Japanese Orthopaedic Association (mJOA) Score
Circle oneI. Motor dysfunction score of the upper extremities
0Inability to move hands
1Inability to eat with a spoon but able to move hands
2Inability to button shirt but able to eat with a spoon
3Able to button shirt with great difficulty
4Able to button shirt with slight difficulty
5No dysfunction
Circle oneII. Motor dysfunction score of the lower extremities
0Complete loss of motor and sensory function
1Sensory preservation without ability to move legs
2Able to move legs but unable to walk
3Able to walk on flat floor with a walking aid (i.e., cane or crutch)
4Able to walk up and/or down stairs with hand rail
5Moderate to significant lack of stability but able to walk up and/or down stairs without hand rail
6Mild lack of stability but walk unaided with smooth reciprocation
7No dysfunction
Circle oneIII. Sensation
0Complete loss of hand sensation
1Severe sensory loss or pain
2Mild sensory loss
3No sensory loss
Circle oneIV. Sphincter dysfunction
0Inability to urinate voluntarily
1Marked difficulty with micturition
2Mild to moderate difficulty with micturition
3Normal micturition
Mild myelopathymJOA from 15 to 17
Moderate myelopathymJOA from 12 to 14
Severe myelopathymJOA from 0 to 11
Table A2. Numeric Pain Rating Scale (NPRS) [257].
Table A2. Numeric Pain Rating Scale (NPRS) [257].
Pain Numeric Rating Scale
1. On a scale of 0 to 10, with 0 being no pain at all and 10 being the worst pain imaginable, how would you rate your pain RIGHT NOW.
012345678910
No Pain Worst Pain Imaginable
2. On the same scale, how would you rate your USUAL level of pain during the last week.
012345678910
No Pain Worst Pain Imaginable
3. On the same scale, how would you rate your BEST level of pain during the last week.
012345678910
No Pain Worst Pain Imaginable
4. On the same scale, how would you rate your WORST level of pain during the last week.
012345678910
No Pain Worst Pain Imaginable
Table A3. Neck Disability Index (NDI) [258].
Table A3. Neck Disability Index (NDI) [258].
Neck Disability Index
Please answer every section and mark in each section only the one box that applies to you.
Section 1: Pain Intensity
I have no pain at the moment
The pain is very mild at the moment
The pain is moderate at the moment
The pain is fairly severe at the moment
The pain is very severe at the moment
The pain is the worst imaginable at the moment
Section 2: Personal Care (Washing, Dressing, etc.)
I can look after myself normally without causing extra pain
I can look after myself normally but it causes extra pain
It is painful to look after myself and I am slow and careful
I need some help but can manage most of my personal care
I need help every day in most aspects of self care
I do not get dressed. I wash with difficulty and stay in bed
Section 3: Lifting
I can lift heavy weights without extra pain
I can lift heavy weights but it gives extra pain
Pain prevents me lifting heavy weights off the floor, but I can manage if they are conveniently placed, for example on a table
Pain prevents me from lifting heavy weights but I can manage light to medium weights if they are conveniently positioned
I can only lift very light weights
I cannot lift or carry anything
Section 4: Reading
I can read as much as I want to with no pain in my neck
I can read as much as I want to with slight pain in my neck
I can read as much as I want with moderate pain in my neck
I can’t read as much as I want because of moderate pain in my neck
I can hardly read at all because of severe pain in my neck
I cannot read at all
Section 5: Headaches
I have no headaches at all
I have slight headaches, which come infrequently
I have moderate headaches, which come infrequently
I have moderate headaches, which come frequently
I have severe headaches, which come frequently
I have headaches almost all the time
Section 6: Concentration
I can concentrate fully when I want to with no difficulty
I can concentrate fully when I want to with slight difficulty
I have a fair degree of difficulty in concentrating when I want to
I have a lot of difficulty in concentrating when I want to
I have a great deal of difficulty in concentrating when I want to
I cannot concentrate at all
Section 7: Work
I can do as much work as I want to
I can only do my usual work, but no more
I can do most of my usual work, but no more
I cannot do my usual work
I can hardly do any work at all
I can’t do any work at all
Section 8: Driving
I can drive my car without any neck pain
I can drive my car as long as I want with slight pain in my neck
I can drive my car as long as I want with moderate pain in my neck
I can’t drive my car as long as I want because of moderate pain in my neck
I can hardly drive at all because of severe pain in my neck
I can’t drive my car at all
Section 9: Sleeping
I have no trouble sleeping
My sleep is slightly disturbed (less than 1 h sleepless)
My sleep is mildly disturbed (1–2 h sleepless)
My sleep is moderately disturbed (2–3 h sleepless)
My sleep is greatly disturbed (3–5 h sleepless)
My sleep is completely disturbed (5–7 h sleepless)
Section 10: Recreation
I am able to engage in all my recreation activities with no neck pain at all
I am able to engage in all my recreation activities, with some pain in my neck
I am able to engage in most, but not all of my usual recreation activities because of pain in my neck
I am able to engage in a few of my usual recreation activities because of pain in my neck
I can hardly do any recreation activities because of pain in my neck
I can’t do any recreation activities at all
Score:___/150 Transform to percentage score x 100 = %points
Scoring: For each section the total possible score is 5: if the first statement is marked the section score = 0, if the last statement is marked it = 5. If all ten sections are completed the score is calculated as follows:
Example: 16 (total scored)50 (total possible score) x 100 = 32%
If one section is missed or not applicable the score is calculated:
Example: 16 (total scored)
45 (total possible score) x 100 = 35.5%
Minimum Detectable Change (90% confidence): 5 points or 10 %points
Table A4. EQ-5D [259].
Table A4. EQ-5D [259].
EQ-5D
By placing a checkmark in one box in each group below, please indicate which statements best describe your own health state today.
Mobility
I have no problems in walking about
I have some problems in walking about
I am confined to bed
Self-Care
I have no problems with self-care
I have some problems washing or dressing myself
I am unable to wash or dress myself
Usual Activities (e.g., work, study, housework, family or leisure activities)
I have no problems with performing my usual activities
I have some problems with performing my usual activities
I am unable to perform my usual activities
Pain/Discomfort
I have no pain or discomfort
I have moderate pain or discomfort
I have extreme pain or discomfort
Anxiety/Depression
I am not anxious or depressed
I am moderately anxious or depressed
I am extremely anxious or depressed
Table A5. Nurick Grading System [260].
Table A5. Nurick Grading System [260].
Nurick Grading System
Grade.Definition
0Signs or symptoms of root involvement, but without evidence of spinal cord disease.
ISigns of spinal cord disease, but no walking difficulty.
IISlight difficulty in walking that does not prevent full- time employment.
IIIWalking difficulty that prevents full-time employment or the ability to do all housework but is not so severe as to require help from another person to ambulate.
IVAble to walk only with help from another person or with the aid of a frame.
VBedridden or chairbound.
Table A6. European Myelopathy Score [49].
Table A6. European Myelopathy Score [49].
European Myelopathy Score
Upper motor neuron
1Unable to walk, wheelchair
Gait function
2Walking on a flat ground only with cane or aid
3Climbing stairs only with aid
4Gait clumsy, but no aid necessary
5Normal walking and climbing stairs
Upper motor neuron
1Retention, no control over bladder and/or bowel function
Bladder and bowel function
2Inadequate micturition and urinary frequency
3Normal bladder and bowel function
Lower motor neuron
1Handwriting and eating with knife and fork impossible
Hand function
2Handwriting and eating with knife and fork impaired
3Handwriting, tying shoe laces or a tie clumsy
4Normal handwriting
Posterior column
1Getting dressed only with aid
Proprioception and coordination
2Getting dressed clumsily and slowly
3Getting dressed normally
Paraesthesia/pain
1Invalidity due to pain
2Endurable paraesthesia and pain
3No paraesthesia and pain
Normal function17–18
Grade 113–16
Grade 29–12
Grade 35–8
Table A7. Cooper Myelopathy Scale [261].
Table A7. Cooper Myelopathy Scale [261].
Cooper Myelopathy Scale
Upper extremity function
Grade 0Intact
Grade 1Sensory symptoms only
Grade 2Mild motor deficit with some functional impairment
Grade 3Major functional impairment in at least one upper extremity but upper extremities useful for simple tasks
Grade 4No movement or flicker of movement in upper extremities; no useful function
Lower extremity function
Grade 0Intact
Grade 1Walks independently but not normally
Grade 2Walks but needs cane or walker
Grade 3Stands but cannot walk
Grade 4Slight movement but cannot walk or stand
Grade 5Paralysis

Appendix B. Database Search Strategy

  • EBM Reviews—ACP Journal Club 1991 to November 2021
  • Embase 1974 to 3 December 2021
  • MEDLINE(R) All including Epub Ahead of Print, In-Process and Other Non-Indexed Citations, Daily and Versions(R) 1946-current
Figure A1. Database search strategy.
Figure A1. Database search strategy.
Biomedicines 10 02621 g0a1

Appendix C. Article Study Characteristics

Table A8. Study characteristics of articles deemed eligible for inclusion by search strategy [60,104,105,108,122,123,124,125,126,127,128,129,130,131,132,133,134,135,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,183,184,185,187,188].
Table A8. Study characteristics of articles deemed eligible for inclusion by search strategy [60,104,105,108,122,123,124,125,126,127,128,129,130,131,132,133,134,135,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,183,184,185,187,188].
No.Author(s)YearTitleStudy DesignFollow-Up Period (Months)SubjectsqMRI TechniqueqMRI Parameters Tested
1Maki, Satoshi; Koda, Masao; Kitamura, Mitsuhiro; Inada, Taigo; Kamiya, Koshiro; Ota, Mitsutoshi; Iijima, Yasushi; Saito, Junya; Masuda, Yoshitada; Matsumoto, Koji; Kojima, Masatoshi; Obata, Takayuki; Takahashi, Kazuhisa; Yamazaki, Masashi; Furuya, Takeo2017Diffusion tensor imaging can predict surgical outcomes of patients with cervical compression myelopathyProspective Longitudinal6DCM = 26DTIFA, MD
2Bhosale, Sunil; Ingale, Pramod; Srivastava, Sudhir; Marathe, Nandan; Bhide, Prajakta2019Diffusion tensor imaging as an additional postoperative prognostic predictor factor in cervical myelopathy patients: An observational studyProspective Longitudinal3DCM = 30DTIFA, MD
3Song, Ting; Chen, Wen-Jun; Huang, Jian-Wei; Cai, Ming-Jin; Dong, Tian-Fa; Li, Tang-Sheng; Yang, Bo; Zhao, Hong-Pu2011Diffusion tensor imaging in the cervical spinal cordProspective Longitudinal6DCM = 53 Healthy Controls = 20DTIFA, ADC
4Severino, Rocco; Nouri, Aria; Tessitore, Enrico2020Degenerative cervical myelopathy: How to identify the best responders to surgery?Prospective Longitudinal12DCM = 36DTIFA
5Nukala, Monika; Abraham, Jini; Khandige, Ganesh; Shetty, Bharath K.; Rao, Arindam pol arjun2019Efficacy of diffusion tensor imaging in identification of degenerative cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 50DTIFA, ADC
6Ulubaba, Hilal Er; Saglik, Semih; Yildirim, Ismail Okan; Durak, Mehmet Akif2021Effectiveness of Diffusion Tensor Imaging in Determining Cervical Spondylotic MyelopathyProspective Cross-sectionalN/ADCM = 54DTIFA, ADC
7Tian, Xiaonan; Zhang, Li; Zhang, Xuesong; Meng, Linghui; Li, Xiaona2021Correlations between preoperative diffusion tensor imaging and surgical outcome in patients with cervical spondylotic myelopathyRetrospective Longitudinal12DCM = 95DTIFA, ADC
8Iwasaki, Motoyuki; Yokohama, Takumi; Oura, Daisuke; Furuya, Shou; Niiya, Yoshimasa; Okuaki, Tomoyuki2019Decreased Value of Highly Accurate Fractional Anisotropy Using 3-Tesla ZOOM Diffusion Tensor Imaging After Decompressive Surgery in Patients with Cervical Spondylotic Myelopathy: Aligned Fibers EffectProspective Longitudinal6DCM = 26Healthy Controls = 12DTIFA
9Toktas, Zafer Orkun; Kilic, Turker; Konya, Deniz; Tanrikulu, Bahattin; Koban, Orkun2016Diffusion tensor imaging of cervical spinal cord: A quantitative diagnostic tool in cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 21DTIFA, ADC
10Ellingson, Benjamin M.; Salamon, Noriko; Grinstead, John W.; Holly, Langston T.2014Diffusion tensor imaging predicts functional impairment in mild-to-moderate cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 48Healthy Controls = 9DTIFA, ADC, MD
11Han, X.; Ma, X.; Li, D.; Wang, J.; Jiang, W.; Cheng, X.; Li, G.; Guo, H.; Tian, W.2020The Evaluation and Prediction of Laminoplasty Surgery Outcome in Patients with Degenerative Cervical Myelopathy Using Diffusion Tensor MRIProspective Longitudinal6DCM = 55Healthy Controls = 20DTIFA, MD
12Guo, Xing; Yang, Xiaotian; Chen, Xukang; Zhao, Rui; Song, Yingchao; Liang, Meng; Sun, Haoran; Xue, Yuan2021Enhanced Information Flow From Cerebellum to Secondary Visual Cortices Leads to Better Surgery Outcome in Degenerative Cervical Myelopathy Patients: A Stochastic Dynamic Causal Modeling Study With Functional Magnetic Resonance ImagingProspective Longitudinal6DCM = 27Healthy Controls = 11fMRI (BOLD)Effective connectivity (EC)
13Rajasekaran, S.; Kanna, Rishi M.; Chittode, Vishnuprasath S.; Maheswaran, Anupama; Aiyer, Siddharth N.; Shetty, Ajoy P.2017Efficacy of Diffusion Tensor Imaging Indices in Assessing Postoperative Neural Recovery in Cervical Spondylotic MyelopathyProspective Longitudinal12DCM = 26DTIADC
14Liu, Xiaojia; Qian, Wenshu; Jin, Richu; Li, Xiang; Luk, Keith Dk; Wu, Ed X.; Hu, Yong2016Amplitude of Low Frequency Fluctuation (ALFF) in the Cervical Spinal Cord with Stenosis: A Resting State fMRI StudyProspective Cross-sectionalN/ADCM = 18Healthy Controls = 25fMRI (BOLD)Amplitude of low frequency fluctuation (ALFF)
15Cui, Jiao-Long; Li, Xiang; Chan, Tin-Yan; Mak, Kin-Cheung; Luk, Keith Dip-Kei; Hu, Yong2015Quantitative assessment of column-specific degeneration in cervical spondylotic myelopathy based on diffusion tensor tractographyProspective Cross-sectionalN/ADCM = 23Healthy Controls = 20DTIFA, MD
16Nischal, Neha; Tripathi, Shalini; Singh, Jatinder Pal2020Quantitative Evaluation of the Diffusion Tensor Imaging Matrix Parameters and the Subsequent Correlation with the Clinical Assessment of Disease Severity in Cervical Spondylotic MyelopathyProspective Cross-sectionalN/ADCM = 52DTIFA, ADC
17Peng, Xinji; Tan, Yongming; He, Laichang; Ou, Yangtao2020Alterations of functional connectivity between thalamus and cortex before and after decompression in cervical spondylotic myelopathy patients: A resting-state functional MRI studyProspective Longitudinal3DCM = 43Healthy Controls = 43fMRI (BOLD)BOLD signal
18Tan, Yongming; Zhou, Fuqing; Liu, Zhili; Wu, Lin; Zeng, Xianjun; Gong, Honghan; He, Laichang2016Alteration of cerebral regional homogeneity within sensorimotor network in patients with cervical spondylotic myelopathy after spinal cord decompression: a resting-state functional MRI studyProspective Longitudinal3DCM = 21Healthy Controls = 21fMRI (BOLD)Regional homogeneity (ReHo)
19Kowalczyk, Izabela; Bartha, Robert; Duggal, Neil2012Proton magnetic resonance spectroscopy of the motor cortex in cervical myelopathyProspective Cross-sectionalN/ADCM = 24Healthy Controls = 11MRSN-acetylaspartate/creatine
20Lee, Seungbo; Chung, Tae-Sub; Kim, Sungjun; Yoo, Yeon Hwa; Yoon, Choon-Sik; Lee, Young Han; Suh, Jin-Suck; Jeong, Eun-Kee; Kim, In Seong; Park, Jung Hyun2015Accuracy of diffusion tensor imaging for diagnosing cervical spondylotic myelopathy in patients showing spinal cord compressionProspective Cross-sectionalN/ADCM = 33DTIFA, MD
21Wang, K.Y.; Idowu, O.; Orman, G.; Izbudak, I.; Thompson, C.B.; Myers, C.; Riley, L.H.; Carrino, J.A.; Flammang, A.; Gilson, W.; Sadowsky, C.L.2017Tract-Specific Diffusion Tensor Imaging in Cervical Spondylotic Myelopathy Before and After Decompressive Spinal Surgery: Preliminary ResultsProspective Longitudinal6DCM = 4Healthy Controls = 5DTIFA, MD
22Shabani, Saman; Kaushal, Mayank; Budde, Matthew; Schmit, Brian; Wang, Marjorie C.; Kurpad, Shekar2019Comparison between quantitative measurements of diffusion tensor imaging and T2 signal intensity in a large series of cervical spondylotic myelopathy patients for assessment of disease severity and prognostication of recoveryProspective Longitudinal24DCM = 46DTIFA
23Duggal, N.; Rabin, D.; Bartha, R.; Barry, R.L.; Gati, J.S.; Kowalczyk, I.; Fink, M.2010Brain reorganization in patients with spinal cord compression evaluated using fMRIProspective Longitudinal6DCM = 12Healthy Controls = 10fMRI (BOLD)Volume of Activation (VOA)
24Jurova, Barbora; Mechl, Marek; Kerkovsky, Milos; Sprlakova-Pukova, Andrea; Kadanka, Zdenek; Nemec, Martin; Bednarik, Josef; Kovalova, Ivana; Dusek, Ladislav2017Spinal Cord MR Diffusion Properties in Patients with Degenerative Cervical Cord CompressionProspective Cross-sectionalN/ADCM = 130Healthy Controls = 71DTIFA, ADC
25Kara, Batuhan; Celik, Azim; Karadereler, Selhan; Ulusoy, Levent; Ganiyusufoglu, Kursat; Onat, Levent; Mutlu, Ayhan; Ornek, Ibrahim; Sirvanci, Mustafa; Hamzaoglu, Azmi2011The role of DTI in early detection of cervical spondylotic myelopathy: a preliminary study with 3-T MRIProspective Cross-sectionalN/ADCM = 16DTIFA, ADC
26Maki, Satoshi; Koda, Masao; Ota, Mitsutoshi; Oikawa, Yoshihiro; Kamiya, Koshiro; Inada, Taigo; Furuya, Takeo; Takahashi, Kazuhisa; Masuda, Yoshitada; Matsumoto, Koji; Kojima, Masatoshi; Obata, Takayuki; Yamazaki, Masashi2018Reduced Field-of-View Diffusion Tensor Imaging of the Spinal Cord Shows Motor Dysfunction of the Lower Extremities in Patients with Cervical Compression MyelopathyProspective Cross-sectionalN/ADCM = 20Healthy Controls = 10DTIFA
27Hassan, Talaat Ahmed Abd El Hameed; Assad, Ramy Edward; Belal, Shaimaa Atef2019MR diffusion tensor imaging of the spinal cord: can it help in early detection of cervical spondylotic myelopathy and assessment of its severity?Prospective Cross-sectionalN/ADCM = 30DTIFA
28Cloney, Michael Brendan; Smith, Zachary A.; Weber, Kenneth A.; Parrish, Todd B.2018Quantitative Magnetization Transfer MRI Measurements of the Anterior Spinal Cord Region are Associated with Clinical Outcomes in Cervical Spondylotic MyelopathyProspective Cross-sectionalN/ADCM = 7Healthy Controls = 7MTMTR
29Salamon, Noriko; Woodworth, Davis C.; Holly, Langston T.; Ellingson, Benjamin M.2018Resting-State Functional Magnetic Resonance Imaging Connectivity of the Brain Is Associated with Altered Sensorimotor Function in Patients with Cervical SpondylosisProspective Cross-sectionalN/ADCM = 24Healthy Controls = 17fMRI (BOLD)Functional Connectivity (FC)
30Wang, Chencai; Salamon, Noriko; Laiwalla, Azim; Holly, Langston T.; Ellingson, Benjamin M.; Islam, Sabah2021Supraspinal functional and structural plasticity in patients undergoing surgery for degenerative cervical myelopathyProspective Longitudinal3DCM = 19Healthy Controls = 16fMRI (BOLD)Functional Connectivity (FC)
31Baucher, G.; Rasoanandrianina, H.; Levy, S.; Pini, L.; Troude, L.; Roche, P. H.; Callot, V.2021T1 Mapping for Microstructural Assessment of the Cervical Spinal Cord in the Evaluation of Patients with Degenerative Cervical MyelopathyProspective Cross-sectionalN/ADCM = 20Healthy Controls = 10Quantitative T1T1
32Banaszek, Anna; Bladowska, Joanna; Szewczyk, Pawel; Podgorski, Przemyslaw; Sasiadek, Marek2014Usefulness of diffusion tensor MR imaging in the assessment of intramedullary changes of the cervical spinal cord in different stages of degenerative spine diseaseProspective Cross-sectionalN/ADCM = 132Healthy Controls = 25DTIFA, ADC
33Ellingson, Benjamin M.; Salamon, Noriko; Hardy, Anthony J.; Holly, Langston T.2015Prediction of Neurological Impairment in Cervical Spondylotic Myelopathy using a Combination of Diffusion MRI and Proton MR SpectroscopyProspective Cross-sectionalN/ADCM = 27Healthy Controls = 11DTI, MRSFA, MD, Cho/NAA (Choline/N-acetylaspartate)
34Salamon, N.; Ellingson, B.M.; Nagarajan, R.; Gebara, N.; Thomas, A.; Holly, L.T.2013Proton magnetic resonance spectroscopy of human cervical spondylosis at 3TProspective Cross-sectionalN/ADCM = 21Healthy Controls = 11MRSNAA (N-acetylaspartate), Cho (choline), Myo-I (myo-inositol) ratio with Cr (creatine)
35Chen, Zhao; Zhao, Rui; Wang, Qiu; Yu, Chunshui; Li, Fengtan; Liang, Meng; Zong, Yaqi; Zhao, Ying; Xiong, Wuyi; Su, Zhe; Xue, Yuan2020Functional Connectivity Changes of the Visual Cortex in the Cervical Spondylotic Myelopathy Patients: A Resting-State fMRI StudyProspective Longitudinal3DCM = 30Healthy Controls = 20fMRI (BOLD)Functional Connectivity (FC)
36Bhagavatula, Indira Devi; Shukla, Dhaval; Sadashiva, Nishanth; Saligoudar, Praveen; Prasad, Chandrajit; Bhat, Dhananjaya I.2016Functional cortical reorganization in cases of cervical spondylotic myelopathy and changes associated with surgeryProspective Longitudinal6DCM = 17Healthy Controls = 12fMRI (BOLD)Volume of Activation (VOA)
37Murphy, Rory K.; Sun, Peng; Han, Rowland H.; Griffin, Kim J.; Wagner, Joanne; Yarbrough, Chester K.; Wright, Neill M.; Dorward, Ian G.; Riew, K. Daniel; Kelly, Michael P.; Santiago, Paul; Zebala, Lukas P.; Trinkaus, Kathryn; Ray, Wilson Z.; Song, Sheng-Kwei2018Fractional anisotropy to quantify cervical spondylotic myelopathy severityProspective Cross-sectionalN/ADCM = 14Healthy Controls = 7DTIFA
38Takenaka, Shota; Kan, Shigeyuki; Seymour, Ben; Makino, Takahiro; Sakai, Yusuke; Kushioka, Junichi; Tanaka, Hisashi; Watanabe, Yoshiyuki; Shibata, Masahiko; Yoshikawa, Hideki; Kaito, Takashi2020Resting-state Amplitude of Low-frequency Fluctuation is a Potentially Useful Prognostic Functional Biomarker in Cervical MyelopathyProspective Longitudinal6DCM = 28Healthy Controls = 28fMRI (BOLD)Amplitude of low frequency fluctuation (ALFF)
39Cui, Libin; Chen, Xueming; Liu, Yadong; Zhang, Yanjun; Kong, Chao; Guan, Yun2019Changes in diffusion tensor imaging indices of the lumbosacral enlargement correlate with cervical spinal cord changes and clinical assessment in patients with cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 40Healthy Controls = 42DTIFA, ADC
40Holly, Langston T.; Wang, Chencai; Salamon, Noriko; Woodworth, Davis C.; Ellingson, Benjamin M.2019Neck disability in patients with cervical spondylosis is associated with altered brain functional connectivityProspective Cross-sectionalN/ADCM = 36Healthy Controls = 17fMRI (BOLD)Functional Connectivity (FC)
41Grabher, Patrick; David, Gergely; Mohammadi, Siawoosh; Freund, Patrick2017Neurodegeneration in the Spinal Ventral Horn Prior to Motor Impairment in Cervical Spondylotic MyelopathyProspective Cross-sectionalN/ADCM = 20Healthy Controls = 18DTIMD
42Kerkovsky, M.; Jakubcova, B.; Mechl, M.; Kadanka, Z.; Kadanka Jr, Z.; Nemec, M.; Kovalova, I.; Bednarik, J.2015Multifactorial determination of the spinal cord diffusion properties in patients with cervical spondylotic spinal cord compression: A diffusion tensor imaging studyProspective Cross-sectionalN/ADCM = 130Healthy Controls = 71DTIFA, ADC
43Kowalczyk, I.; Bartha, R.; Duggal, N.2010Proton magnetic resonance spectroscopy of the motor cortex in cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 24Healthy Controls = 11MRSNAA/Cr (N-acetylaspartate/creatine metabolite ratio)
44Taha Ali, Tamer F.; Badawy, Ahmed E.2013Feasibility of 1H-MR Spectroscopy in evaluation of cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 34Healthy Controls = 11MRSNAA/Cr (N-acetylaspartate/creatine metabolite ratio), Cho/Cr (Chloline/creatine ratio)
45Aleksanderek, Izabela K.; Stevens, Todd; Goncalves, Sandy; Bartha, Robert; Duggal, Neil2017Metabolite and functional profile of patients with cervical spondylotic myelopathyProspective Longitudinal6DCM = 28Healthy Controls = 10fMRI (BOLD), MRSVolume of Activation (VOA), NAA/Cr (N-acetylaspartate/creatine metabolite ratio)
46Wen, Chun Yi; Cui, Jiao Long; Liu, Harris S.; Mak, Kin Cheung; Cheung, Wai Yuen; Luk, Keith D.K.; Hu, Yong2014Is diffusion anisotropy a biomarker for disease severity and surgical prognosis of cervical spondylotic myelopathyProspective Longitudinal6 to 24DCM = 45Healthy Controls = 20DTIFA
47Paliwal, Monica; Smith, Zachary A.; Weber, Kenneth A.; Mackey, Sean; Hopkins, Benjamin S.; Dahdaleh, Nader S.; Cantrell, Donald R.; Parrish, Todd D.; Hoggarth, Mark A.; Elliott, James M.; Dhaher, Yasin2020Magnetization Transfer Ratio and Morphometrics of the Spinal Cord Associates with Surgical Recovery in Patients with Degenerative Cervical MyelopathyProspective Longitudinal6DCM = 13Healthy Controls = 9MTMTR
48Martin, Allan R.; De Leener, Benjamin; Cohen-Adad, Julien; Kalsi-Ryan, Sukhvinder; Cadotte, David W.; Wilson, Jefferson R.; Tetreault, Lindsay; Nouri, Aria; Crawley, Adrian; Mikulis, David J.; Ginsberg, Howard; Massicotte, Eric M.; Fehlings, Michael G.2018Monitoring for myelopathic progression with multiparametric quantitative MRIProspective Longitudinal12DCM = 26DTI, MTFA, MTR
49Chen, Xueming; Kong, Chao; Feng, Shiqing; Guan, Hua; Yu, Zhenshan; Cui, Libin; Wang, Yanhui2016Magnetic resonance diffusion tensor imaging of cervical spinal cord and lumbosacral enlargement in patients with cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 10Healthy Controls = 10DTIFA, ADC
50Suleiman, Linda I.; Rosenthal, Brett D.; Bhatt, Surabhi A.; Hsu, Wellington K.; Patel, Alpesh A.; Parrish, Todd B.; Savage, Jason W.; Weber, Kenneth A.2018High-resolution magnetization transfer MRI in patients with cervical spondylotic myelopathyProspective Cross-sectionalN/ADCM = 10Healthy Controls = 7MTMTR
51Nagashima, Hideki; Nanjo, Yoshiro; Teshima, Ryota; Morio, Yasuo; Meshitsuka, Shunsuke; Yamane, Koji2010High-resolution nuclear magnetic resonance spectroscopic study of metabolites in the cerebrospinal fluid of patients with cervical myelopathy and lumbar radiculopathyProspective Cross-sectionalN/ADCM = 30Healthy Controls = 10MRSLactate, alanine, acetate, glutamate, pyruvate, citrate
52Su, Qian; Zhao, Rui; Guo, Xing; Wang, ShuoWen; Tu, HaoYang; Yang, Fan2021Identification and Therapeutic Outcome Prediction of Cervical Spondylotic Myelopathy Based on the Functional Connectivity From Resting-State Functional MRI Data: A Preliminary Machine Learning StudyRetrospective Longitudinal6DCM = 53Healthy Controls = 47fMRI (BOLD)Functional Connectivity (FC)
53Yang, Young-Mi; Oh, Jae-Keun; Song, Ji-Sun; Yoo, Woo-Kyoung; Yoo, Je Hyun; Kwak, Yoon Hae; Kim, Seok Woo2017The functional relevance of diffusion tensor imaging in comparison to conventional MRI in patients with cervical compressive myelopathyProspective Cross-sectionalN/ADCM = 20DTIFA, ADC
54Zhang, Meng-Ze; Liu, Jian-Fang; Jin, Dan; Wang, Chun-Jie; Zhao, Qiang; Lang, Ning; Yuan, Hui-Shu; Ou-Yang, Han-Qiang; Liu, Xiao-Guang; Liu, Zhong-Jun; Jiang, Liang; Zhang, Xian-Chang2021Utility of Advanced DWI in the Detection of Spinal Cord Microstructural Alterations and Assessment of Neurologic Function in Cervical Spondylotic Myelopathy PatientsRetrospective Longitudinal3DCM = 48Healthy Controls = 36DTIFA
55Xiangshui, M.; Xiangjun, C.; Xiaoming, Z.; Qingshi, Z.; Yi, C.; Chuanqiang, Q.; Xiangxing, M.; Chuanfu, L.; Jinwen, H.20103 T magnetic resonance diffusion tensor imaging and fibre tracking in cervical myelopathyProspective Cross-sectionalN/ADCM = 84Healthy Controls = 21DTIFA, ADC
56He, Zhen; Wang, Nan; Kang, Liqing; Cui, Jiaolong; Wan, Yeda2020Analysis of pathological parameters of cervical spondylotic myelopathy using magnetic resonance imagingProspective Cross-sectionalN/ADCM = 31Healthy Controls = 8DTIFA
57Mamata, Hatsuho; Jolesz, Ferenc A.; Maier, Stephan E.2005Apparent diffusion coefficient and fractional anisotropy in spinal cord: age and cervical spondylosis-related changesProspective Cross-sectionalN/ADCM = 79Healthy Controls = 11DTIFA, ADC
58Zheng, Weipeng; Chen, Haoyi; Wang, Ning; Jiang, Xin; Liang, YingJie; Xiao, Wende; Zhong, Bofu; Ju, Hongbin; Luo, Junnan; Wen, Shifeng; Xiong, Weifeng2018Application of Diffusion Tensor Imaging Cutoff Value to Evaluate the Severity and Postoperative Neurologic Recovery of Cervical Spondylotic MyelopathyRetrospective Longitudinal12 to 24DCM = 61DTIADC, MD
59Kanchiku, T.; Imajo, Y.; Suzuki, H.; Yoshida, Y.; Nishida, N.; Taguchi, T.; Suetomi, Y.; Nishijima, S.2016Application of diffusion tensor imaging for the diagnosis of segmental level of dysfunction in cervical spondylotic myelopathyRetrospective Cross-sectionalN/ADCM = 10Healthy Controls = 11DTIFA, ADC
60Uda, Takehiro; Takami, Toshihiro; Tsuyuguchi, Naohiro; Sakamoto, Shinichi; Yamagata, Toru; Ikeda, Hidetoshi; Nagata, Takashi; Ohata, Kenji2013Assessment of cervical spondylotic myelopathy using diffusion tensor magnetic resonance imaging parameter at 3.0 teslaProspective Cross-sectionalN/ADCM = 26Healthy Controls = 30DTIFA, MD
61Rajasekaran, S.; Kanna, Rishi M.; Balamurali, Gopalakrishnan; Shetty, Ajoy Prasad; Yerramshetty, Janardhan S.; Chittode, Vishnuprasath S.2014The assessment of neuronal status in normal and cervical spondylotic myelopathy using diffusion tensor imagingProspective Cross-sectionalN/ADCM = 35Healthy Controls = 40DTIADC
62Maier, Ilko L; Hofer, Sabine; Eggert, Eva; Schregel, Katharina; Psychogios, Marios-Nikos; Frahm, Jens; Bähr, Mathias; Liman, Jan2020T1 Mapping Quantifies Spinal Cord Compression in Patients With Various Degrees of Cervical Spinal Canal StenosisProspective Cross-sectionalN/ADCM = 31Healthy Controls = 10Quantitative T1T1
63Albistegui-Dubois, Richard; Marehbian, Jonathan; Newton, Jennifer M.; Dong, Yun; Holly, Langston T.; Yan, Xiaohong; Dobkin, Bruce H.2008Compensatory cerebral adaptations before and evolving changes after surgical decompression in cervical spondylotic myelopathy: Laboratory investigationProspective Longitudinal6DCM = 8Healthy Controls = 6fMRI (BOLD)Volume of Activation (VOA)
64Hori, Masaaki; Fukunaga, Issei; Masutani, Yoshitaka; Nakanishi, Atsushi; Shimoji, Keigo; Kamagata, Koji; Asahi, Koichi; Hamasaki, Nozomi; Suzuki, Yuriko; Aoki, Shigeki2012New diffusion metrics for spondylotic myelopathy at an early clinical stageProspective Cross-sectionalN/ADCM = 50DTIFA, ADC
65Vedantam, Aditya; Rao, Avinash; Kurpad, Shekar N.; Jirjis, Michael B.; Eckardt, Gerald; Schmit, Brian D.; Wang, Marjorie C.2017Diffusion Tensor Imaging Correlates with Short-Term Myelopathy Outcome in Patients with Cervical Spondylotic MyelopathyProspective Longitudinal3DCM = 27DTIFA
66Wang, Kun; Chen, Zhi; Shen, Hongxing; Zhang, Fan; Song, Qingxin; Hou, Canglong; Tang, Yixing; Wang, Jun; Chen, Shiyue; Bian, Yun; Hao, Qiang2017Evaluation of DTI Parameter Ratios and Diffusion Tensor Tractography Grading in the Diagnosis and Prognosis Prediction of Cervical Spondylotic MyelopathyProspective Longitudinal12DCM = 93Healthy Controls = 36DTIFA, ADC
67Sato, T.; Horikoshi, T.; Watanabe, A.; Uchida, M.; Ishigame, K.; Araki, T.; Kinouchi, H.2012Evaluation of cervical myelopathy using apparent diffusion coefficient measured by diffusion-weighted imagingProspective Longitudinal6DCM = 66DTIADC
68Takenaka, Shota; Kan, Shigeyuki; Seymour, Ben; Makino, Takahiro; Sakai, Yusuke; Kushioka, Junichi; Tanaka, Hisashi; Watanabe, Yoshiyuki; Shibata, Masahiko; Yoshikawa, Hideki; Kaito, Takashi2019Towards prognostic functional brain biomarkers for cervical myelopathy: A resting-state fMRI studyProspective Longitudinal6DCM = 28Healthy Controls = 28fMRI (BOLD)Functional Connectivity (FC)

References

  1. The Lancet Neurology. A focus on patient outcomes in cervical myelopathy. Lancet Neurol. 2019, 18, 615. [Google Scholar] [CrossRef] [Green Version]
  2. Tracy, J.A.; Bartleson, J. Cervical spondylotic myelopathy. Neurology 2010, 16, 176–187. [Google Scholar] [CrossRef] [PubMed]
  3. Ide, M.; Ogata, H.; Tokuhiro, A.; Takechi, H. Spinal cord injuries in Okayama Prefecture: An epidemiological study 88–89. J. UOEH 1993, 15, 209–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. McKinley, W.O.; Seel, R.T.; Hardman, J.T. Nontraumatic spinal cord injury: Incidence, epidemiology, and functional outcome. Arch. Phys. Med. Rehabil. 1999, 80, 619–623. [Google Scholar] [CrossRef]
  5. New, P.W. Functional outcomes and disability after nontraumatic spinal cord injury rehabilitation: Results from a retrospective study. Arch. Phys. Med. Rehabil. 2005, 86, 250–261. [Google Scholar] [CrossRef] [PubMed]
  6. New, P.W.; Rawicki, H.B.; Bailey, M.J. Nontraumatic spinal cord injury: Demographic characteristics and complications. Arch. Phys. Med. Rehabil. 2002, 83, 996–1001. [Google Scholar] [CrossRef] [PubMed]
  7. New, P.W.; Cripps, R.A.; Lee, B.B. Global maps of non-traumatic spinal cord injury epidemiology: Towards a living data repository. Spinal Cord 2014, 52, 97–109. [Google Scholar] [CrossRef]
  8. Biering-Sørensen, F.; Pedersen, V.; Clausen, S. Epidemiology of spinal cord lesions in Denmark. Spinal Cord 1990, 28, 105–118. [Google Scholar] [CrossRef] [Green Version]
  9. Ronen, J.; Goldin, D.; Bluvshtein, V.; Fishel, B.; Gelernter, I.; Catz, A. Survival after nontraumatic spinal cord lesions in Israel. Arch. Phys. Med. Rehabil. 2004, 85, 1499–1502. [Google Scholar] [CrossRef] [PubMed]
  10. Catz, A.; Goldin, D.; Fishel, B.; Ronen, J.; Bluvshtein, V.; Gelernter, I. Recovery of neurologic function following nontraumatic spinal cord lesions in Israel. Spine 2004, 29, 2278–2282. [Google Scholar] [CrossRef]
  11. Citterio, A.; Franceschini, M.; Spizzichino, L.; Reggio, A.; Rossi, B.; Stampacchia, G.; Mielolesioni, G.I.S.E. Nontraumatic spinal cord injury: An Italian survey. Arch. Phys. Med. Rehabil. 2004, 85, 1483–1487. [Google Scholar] [CrossRef] [PubMed]
  12. Scivoletto, G.; Farchi, S.; Laurenza, L.; Molinari, M. Traumatic and non-traumatic spinal cord lesions: An Italian comparison of neurological and functional outcomes. Spinal Cord 2011, 49, 391–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Schönherr, M.; Groothoff, J.; Mulder, G.; Eisma, W. Rehabilitation of patients with spinal cord lesions in The Netherlands: An epidemiological study. Spinal Cord 1996, 34, 679–683. [Google Scholar] [CrossRef] [PubMed]
  14. Buchan, A.; Fulford, G.; Harris, P.; Jellinek, E.; Kerr, W.; Kirkland, I.; Newsam, J.; Stark, G. A preliminary survey of the incidence and aetiology of spinal paralysis. Spinal Cord 1972, 10, 23–28. [Google Scholar] [CrossRef] [PubMed]
  15. New, P.W.; Farry, A.; Baxter, D.; Noonan, V. Prevalence of non-traumatic spinal cord injury in Victoria, Australia. Spinal Cord 2013, 51, 99–102. [Google Scholar] [CrossRef] [PubMed]
  16. Nouri, A.; Tetreault, L.; Singh, A.; Karadimas, S.K.; Fehlings, M.G. Degenerative cervical myelopathy: Epidemiology, genetics, and pathogenesis. Spine 2015, 40, E675–E693. [Google Scholar] [CrossRef] [Green Version]
  17. Tu, J.; Vargas Castillo, J.; Das, A.; Diwan, A.D. Degenerative Cervical Myelopathy: Insights into Its Pathobiology and Molecular Mechanisms. J. Clin. Med. 2021, 10, 1214. [Google Scholar] [CrossRef] [PubMed]
  18. Badhiwala, J.H.; Witiw, C.D.; Nassiri, F.; Akbar, M.A.; Mansouri, A.; Wilson, J.R.; Fehlings, M.G. Efficacy and safety of surgery for mild degenerative cervical myelopathy: Results of the AOSpine North America and international prospective multicenter studies. Neurosurgery 2019, 84, 890–897. [Google Scholar] [CrossRef] [Green Version]
  19. Badhiwala, J.H.; Wilson, J.R. The natural history of degenerative cervical myelopathy. Neurosurg. Clin. 2018, 29, 21–32. [Google Scholar] [CrossRef]
  20. Nakamura, K.; Kurokawa, T.; Hoshino, Y.; Saita, K.; Takeshita, K.; Kawaguchi, H. Conservative treatment for cervical spondylotic myelopathy: Achievement and sustainability of a level of “no disability”. J. Spinal Disord. 1998, 11, 175–179. [Google Scholar] [CrossRef] [PubMed]
  21. Clarke, E.; Robinson, P.K. Cervical myelopathy: A complication of cervical spondylosis. Brain 1956, 79, 483–510. [Google Scholar] [CrossRef] [PubMed]
  22. Bednarík, J.; Kadanka, Z.; Vohánka, S.; Stejskal, L.; Vlach, O.; Schröder, R. The value of somatosensory-and motor-evoked potentials in predicting and monitoring the effect of therapy in spondylotic cervical myelopathy: Prospective randomized study. Spine 1999, 24, 1593. [Google Scholar] [CrossRef] [PubMed]
  23. Kadanka, Z.; Mareš, M.; Bednarík, J.; Smrcka, V.; Krbec, M.; Stejskal, L.; Chaloupka, R.; Dagmar, S.; Novotný, O.; Urbánek, I. Approaches to spondylotic cervical myelopathy: Conservative versus surgical results in a 3-year follow-up study. Spine 2002, 27, 2205–2210. [Google Scholar] [CrossRef] [PubMed]
  24. Kadaňka, Z.; Bednařík, J.; Novotný, O.; Urbánek, I.; Dušek, L. Cervical spondylotic myelopathy: Conservative versus surgical treatment after 10 years. Eur. Spine J. 2011, 20, 1533–1538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kadaňka, Z.; Bednařík, J.; Voháňka, S.; Vlach, O.; Stejskal, L.; Chaloupka, R.; Filipovičová, D.; Šurelová, D.; Adamová, B.; Novotný, O. Conservative treatment versus surgery in spondylotic cervical myelopathy: A prospective randomised study. Eur. Spine J. 2000, 9, 538–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kadaňka, Z.; Mareš, M.; Bednařík, J.; Smrčka, V.; Krbec, M.; Chaloupka, R.; Dušek, L. Predictive factors for mild forms of spondylotic cervical myelopathy treated conservatively or surgically. Eur. J. Neurol. 2005, 12, 16–24. [Google Scholar] [CrossRef] [PubMed]
  27. Matsumoto, M.; Toyama, Y.; Ishikawa, M.; Chiba, K.; Suzuki, N.; Fujimura, Y. Increased signal intensity of the spinal cord on magnetic resonance images in cervical compressive myelopathy: Does it predict the outcome of conservative treatment? Spine 2000, 25, 677–682. [Google Scholar] [CrossRef] [PubMed]
  28. Sampath, P.; Bendebba, M.; Davis, J.D.; Ducker, T.B. Outcome of patients treated for cervical myelopathy: A prospective, multicenter study with independent clinical review. Spine 2000, 25, 670–676. [Google Scholar] [CrossRef] [PubMed]
  29. Matsumoto, M.; Chiba, K.; Ishikawa, M.; Maruiwa, H.; Fujimura, Y.; Toyama, Y. Relationships between outcomes of conservative treatment and magnetic resonance imaging findings in patients with mild cervical myelopathy caused by soft disc herniations. Spine 2001, 26, 1592–1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Yoshimatsu, H.; Nagata, K.; Goto, H.; Sonoda, K.; Ando, N.; Imoto, H.; Mashima, T.; Takamiya, Y. Conservative treatment for cervical spondylotic myelopathy: Prediction of treatment effects by multivariate analysis. Spine J. 2001, 1, 269–273. [Google Scholar] [CrossRef]
  31. Sumi, M.; Miyamoto, H.; Suzuki, T.; Kaneyama, S.; Kanatani, T.; Uno, K. Prospective cohort study of mild cervical spondylotic myelopathy without surgical treatment. J. Neurosurg. Spine 2012, 16, 8–14. [Google Scholar] [CrossRef] [PubMed]
  32. Shimomura, T.; Sumi, M.; Nishida, K.; Maeno, K.; Tadokoro, K.; Miyamoto, H.; Kurosaka, M.; Doita, M. Prognostic factors for deterioration of patients with cervical spondylotic myelopathy after nonsurgical treatment. Spine 2007, 32, 2474–2479. [Google Scholar] [CrossRef]
  33. Oshima, Y.; Seichi, A.; Takeshita, K.; Chikuda, H.; Ono, T.; Baba, S.; Morii, J.; Oka, H.; Kawaguchi, H.; Nakamura, K. Natural course and prognostic factors in patients with mild cervical spondylotic myelopathy with increased signal intensity on T2-weighted magnetic resonance imaging. Spine 2012, 37, 1909–1913. [Google Scholar] [CrossRef] [PubMed]
  34. Rhee, J.; Tetreault, L.A.; Chapman, J.R.; Wilson, J.R.; Smith, J.S.; Martin, A.R.; Dettori, J.R.; Fehlings, M.G. Nonoperative versus operative management for the treatment degenerative cervical myelopathy: An updated systematic review. Glob. Spine J. 2017, 7, 35S–41S. [Google Scholar] [CrossRef] [Green Version]
  35. Karadimas, S.; Erwin, W.; Ely, C.; Dettori, J.; Fehlings, M. Pathophysiology and natural history of cervical spondylotic myelopathy. Spine 2013, 38, S21–S36. [Google Scholar] [CrossRef] [PubMed]
  36. Tetreault, L.A.; Karadimas, S.; Wilson, J.R.; Arnold, P.M.; Kurpad, S.; Dettori, J.R.; Fehlings, M.G. The natural history of degenerative cervical myelopathy and the rate of hospitalization following spinal cord injury: An updated systematic review. Glob. Spine J. 2017, 7, 28S–34S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Kalsi-Ryan, S.; Karadimas, S.K.; Fehlings, M.G. Cervical spondylotic myelopathy: The clinical phenomenon and the current pathobiology of an increasingly prevalent and devastating disorder. Neuroscientist 2013, 19, 409–421. [Google Scholar] [CrossRef] [PubMed]
  38. Tetreault, L.; Goldstein, C.L.; Arnold, P.; Harrop, J.; Hilibrand, A.; Nouri, A.; Fehlings, M.G. Degenerative cervical myelopathy: A spectrum of related disorders affecting the aging spine. Neurosurgery 2015, 77, S51–S67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Davies, B.M.; Munro, C.F.; Kotter, M.R. A novel insight into the challenges of diagnosing degenerative cervical myelopathy using web-based symptom Checkers. J. Med. Internet Res. 2019, 21, e10868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Davies, B.M.; Mowforth, O.D.; Smith, E.K.; Kotter, M.R. Degenerative cervical myelopathy. BMJ 2018, 360, k186. [Google Scholar] [CrossRef] [PubMed]
  41. Tetreault, L.; Kopjar, B.; Nouri, A.; Arnold, P.; Barbagallo, G.; Bartels, R.; Qiang, Z.; Singh, A.; Zileli, M.; Vaccaro, A. The modified Japanese Orthopaedic Association scale: Establishing criteria for mild, moderate and severe impairment in patients with degenerative cervical myelopathy. Eur. Spine J. 2017, 26, 78–84. [Google Scholar] [CrossRef] [PubMed]
  42. Harrop, J.S.; Naroji, S.; Maltenfort, M.; Anderson, D.G.; Albert, T.; Ratliff, J.K.; Ponnappan, R.K.; Rihn, J.A.; Smith, H.E.; Hilibrand, A. Cervical myelopathy: A clinical and radiographic evaluation and correlation to cervical spondylotic myelopathy. Spine 2010, 35, 620–624. [Google Scholar] [CrossRef] [PubMed]
  43. Iyer, A.; Azad, T.D.; Tharin, S. Cervical spondylotic myelopathy. Clin. Spine Surg. 2016, 29, 408–414. [Google Scholar] [CrossRef] [PubMed]
  44. Cook, C.E.; Wilhelm, M.; Cook, A.E.; Petrosino, C.; Isaacs, R. Clinical tests for screening and diagnosis of cervical spine myelopathy: A systematic review. J. Manip. Physiol. Ther. 2011, 34, 539–546. [Google Scholar] [CrossRef] [PubMed]
  45. Davies, B.M.; McHugh, M.; Elgheriani, A.; Kolias, A.G.; Tetreault, L.A.; Hutchinson, P.J.; Fehlings, M.G.; Kotter, M.R. Reported outcome measures in degenerative cervical myelopathy: A systematic review. PLoS ONE 2016, 11, e0157263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kopjar, B.; Tetreault, L.; Kalsi-Ryan, S.; Fehlings, M. Psychometric properties of the modified Japanese Orthopaedic Association scale in patients with cervical spondylotic myelopathy. Spine 2015, 40, E23–E28. [Google Scholar] [CrossRef] [Green Version]
  47. Revanappa, K.K.; Rajshekhar, V. Comparison of Nurick grading system and modified Japanese Orthopaedic Association scoring system in evaluation of patients with cervical spondylotic myelopathy. Eur. Spine J. 2011, 20, 1545–1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Vitzthum, H.-E.; Dalitz, K. Analysis of five specific scores for cervical spondylogenic myelopathy. Eur. Spine J. 2007, 16, 2096–2103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Herdmann, J.; Linzbach, M.; Krzan, M.; Dvorak, J.; Bock, W. The European myelopathy score. In Cerebellar Infarct. Midline Tumors. Minimally Invasive Endoscopic Neurosurgery (MIEN); Springer: Berlin/Heidelberg, Germany, 1994; pp. 266–268. [Google Scholar]
  50. Lebl, D.R.; Hughes, A.; Cammisa, F.P., Jr.; O’leary, P.F. Cervical spondylotic myelopathy: Pathophysiology, clinical presentation, and treatment. HSS J. 2011, 7, 170–178. [Google Scholar] [CrossRef] [Green Version]
  51. Singh, A.; Tetreault, L.; Casey, A.; Laing, R.; Statham, P.; Fehlings, M.G. A summary of assessment tools for patients suffering from cervical spondylotic myelopathy: A systematic review on validity, reliability and responsiveness. Eur. Spine J. 2015, 24, 209–228. [Google Scholar] [CrossRef] [PubMed]
  52. Furlan, J.C.; Craven, B.C. Psychometric analysis and critical appraisal of the original, revised, and modified versions of the Japanese Orthopaedic Association score in the assessment of patients with cervical spondylotic myelopathy. Neurosurg. Focus 2016, 40, E6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Zhou, F.; Zhang, Y.; Sun, Y.; Zhang, F.; Pan, S.; Liu, Z. Assessment of the minimum clinically important difference in neurological function and quality of life after surgery in cervical spondylotic myelopathy patients: A prospective cohort study. Eur. Spine J. 2015, 24, 2918–2923. [Google Scholar] [CrossRef] [PubMed]
  54. Singh, A.; Crockard, H. Comparison of seven different scales used to quantify severity of cervical spondylotic myelopathy and post-operative improvement. J. Outcome Meas. 2001, 5, 798–818. [Google Scholar] [PubMed]
  55. Iohom, G. Chapter 11—Clinical Assessment of Postoperative Pain. In Postoperative Pain Management; Shorten, G., Carr, D.B., Harmon, D., Puig, M.M., Browne, J., Eds.; W.B. Saunders: Philadelphia, PA, USA, 2006; pp. 102–108. [Google Scholar]
  56. Balestroni, G.; Bertolotti, G. EuroQol-5D (EQ–5D): An instrument for measuring quality of life. Monaldi Arch. Chest Dis. 2012, 78, 155–159. [Google Scholar] [CrossRef] [PubMed]
  57. Nouri, A.; Martin, A.R.; Mikulis, D.; Fehlings, M.G. Magnetic resonance imaging assessment of degenerative cervical myelopathy: A review of structural changes and measurement techniques. Neurosurg. Focus 2016, 40, E5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Nagata, K.; Kiyonaga, K.; Ohashi, T.; Sagara, M.; Miyazaki, S.; Inoue, A. Clinical value of magnetic resonance imaging for cervical myelopathy. Spine 1990, 15, 1088–1096. [Google Scholar] [CrossRef]
  59. Sun, Q.; Hu, H.; Zhang, Y.; Li, Y.; Chen, L.; Chen, H.; Yuan, W. Do intramedullary spinal cord changes in signal intensity on MRI affect surgical opportunity and approach for cervical myelopathy due to ossification of the posterior longitudinal ligament? Eur. Spine J. 2011, 20, 1466–1473. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, Y.-M.; Yoo, W.-K.; Yoo, J.H.; Kwak, Y.H.; Oh, J.-K.; Song, J.-S.; Kim, S.W. The functional relevance of diffusion tensor imaging in comparison to conventional MRI in patients with cervical compressive myelopathy. Skelet. Radiol. 2017, 46, 1477–1486. [Google Scholar] [CrossRef]
  61. Houser, O.W.; Onofrio, B.M.; Miller, G.M.; Folger, W.N.; Smith, P.L. Cervical spondylotic stenosis and myelopathy: Evaluation with computed tomographic myelography. In Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 1994; Volume 69, pp. 557–563. [Google Scholar]
  62. Takahashi, M.; Yamashita, Y.; Sakamoto, Y.; Kojima, R. Chronic cervical cord compression: Clinical significance of increased signal intensity on MR images. Radiology 1989, 173, 219–224. [Google Scholar] [CrossRef]
  63. Suzuki, A.; Daubs, M.D.; Inoue, H.; Hayashi, T.; Aghdasi, B.; Montgomery, S.R.; Ruangchainikom, M.; Hu, X.; Lee, C.J.; Wang, C.J. Prevalence and motion characteristics of degenerative cervical spondylolisthesis in the symptomatic adult. Spine 2013, 38, E1115–E1120. [Google Scholar] [CrossRef]
  64. Muhle, C.; Metzner, J.; Weinert, D.; Falliner, A.; Brinkmann, G.; Mehdorn, M.H.; Heller, M.; Resnick, D. Classification system based on kinematic MR imaging in cervical spondylitic myelopathy. Am. J. Neuroradiol. 1998, 19, 1763–1771. [Google Scholar] [PubMed]
  65. Kang, Y.; Lee, J.; Koh, Y.; Hur, S.; Kim, S.; Chai, J. New MRI grading system for the cervical canal stenosis. AJR Am. J. Roentgenol. 2011, 197, W134–W140. [Google Scholar] [CrossRef] [PubMed]
  66. Fujiwara, K.; Yonenobu, K.; Hiroshima, K.; Ebara, S.; Yamashita, K.; Ono, K. Morphometry of the cervical spinal cord and its relation to pathology in cases with compression myelopathy. Spine 1988, 13, 1212–1216. [Google Scholar] [CrossRef]
  67. Okada, Y.; Ikata, T.; Yamada, H.; Sakamoto, R.; Katoh, S. Magnetic resonance imaging study on the results of surgery for cervical compression myelopathy. Spine 1993, 18, 2024–2029. [Google Scholar] [CrossRef]
  68. Furlan, J.C.; Kailaya-Vasan, A.; Aarabi, B.; Fehlings, M.G. A novel approach to quantitatively assess posttraumatic cervical spinal canal compromise and spinal cord compression: A multicenter responsiveness study. Spine 2011, 36, 784–793. [Google Scholar] [CrossRef]
  69. Fehlings, M.G.; Rao, S.C.; Tator, C.H.; Skaf, G.; Arnold, P.; Benzel, E.; Dickman, C.; Cuddy, B.; Green, B.; Hitchon, P. The optimal radiologic method for assessing spinal canal compromise and cord compression in patients with cervical spinal cord injury: Part II: Results of a multicenter study. Spine 1999, 24, 605–613. [Google Scholar] [CrossRef]
  70. Nouri, A.; Martin, A.R.; Kato, S.; Kermani, H.R.; Riehm, L.; Fehlings, M.G. The Relationship Between MRI Signal Intensity Changes, Clinical Presentation and Surgical Outcome in Degenerative Cervical Myelopathy: Analysis of a Global Cohort. Spine J. 2017, 17, S133–S134. [Google Scholar] [CrossRef]
  71. Uchida, K.; Nakajima, H.; Takeura, N.; Yayama, T.; Guerrero, A.R.; Yoshida, A.; Sakamoto, T.; Honjoh, K.; Baba, H. Prognostic value of changes in spinal cord signal intensity on magnetic resonance imaging in patients with cervical compressive myelopathy. Spine J. 2014, 14, 1601–1610. [Google Scholar] [CrossRef]
  72. Papadopoulos, C.A.; Karonis, P.; Papagelopoulos, P.J.; Karampekios, S.; Hadjipavlou, A.G. Surgical Decompression for Cervical Spondylotic Myelopathy: Correlation between Operative Outcomes and MRI of the Spinal Cord; SLACK Incorporated: Thorofare, NJ, USA, 2004. [Google Scholar]
  73. de Rota, J.J.F.; Meschian, S.; de Rota, A.F.; Urbano, V.; Baron, M. Cervical spondylotic myelopathy due to chronic compression: The role of signal intensity changes in magnetic resonance images. J. Neurosurg. Spine 2007, 6, 17–22. [Google Scholar]
  74. Mastronardi, L.; Elsawaf, A.; Roperto, R.; Bozzao, A.; Caroli, M.; Ferrante, M.; Ferrante, L. Prognostic relevance of the postoperative evolution of intramedullary spinal cord changes in signal intensity on magnetic resonance imaging after anterior decompression for cervical spondylotic myelopathy. J. Neurosurg. Spine 2007, 7, 615–622. [Google Scholar] [CrossRef]
  75. Yagi, M.; Ninomiya, K.; Kihara, M.; Horiuchi, Y. Long-term surgical outcome and risk factors in patients with cervical myelopathy and a change in signal intensity of intramedullary spinal cord on magnetic resonance imaging. J. Neurosurg. Spine 2010, 12, 59–65. [Google Scholar] [CrossRef] [PubMed]
  76. Yukawa, Y.; Kato, F.; Yoshihara, H.; Yanase, M.; Ito, K. MR T2 image classification in cervical compression myelopathy: Predictor of surgical outcomes. Spine 2007, 32, 1675–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Nouri, A.; Tetreault, L.; Côté, P.; Zamorano, J.J.; Dalzell, K.; Fehlings, M.G. Does magnetic resonance imaging improve the predictive performance of a validated clinical prediction rule developed to evaluate surgical outcome in patients with degenerative cervical myelopathy? Spine 2015, 40, 1092–1100. [Google Scholar] [CrossRef]
  78. Kato, F.; Yukawa, Y.; Suda, K.; Yamagata, M.; Ueta, T. Normal morphology, age-related changes and abnormal findings of the cervical spine. Part II: Magnetic resonance imaging of over 1200 asymptomatic subjects. Eur. Spine J. 2012, 21, 1499–1507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Wessberg, P.; Danielson, B.I.; Willén, J. Comparison of Cobb angles in idiopathic scoliosis on standing radiographs and supine axially loaded MRI. Spine 2006, 31, 3039–3044. [Google Scholar] [CrossRef] [PubMed]
  80. Cowley, P. Neuroimaging of spinal canal stenosis. Magn. Reson. Imaging Clin. 2016, 24, 523–539. [Google Scholar] [CrossRef] [PubMed]
  81. Karpova, A.; Arun, R.; Kalsi-Ryan, S.; Massicotte, E.M.; Kopjar, B.; Fehlings, M.G. Do quantitative magnetic resonance imaging parameters correlate with the clinical presentation and functional outcomes after surgery in cervical spondylotic myelopathy? A prospective multicenter study. Spine 2014, 39, 1488–1497. [Google Scholar] [CrossRef] [PubMed]
  82. Li, F.; Chen, Z.; Zhang, F.; Shen, H.; Hou, T. A meta-analysis showing that high signal intensity on T2-weighted MRI is associated with poor prognosis for patients with cervical spondylotic myelopathy. J. Clin. Neurosci. 2011, 18, 1592–1595. [Google Scholar] [CrossRef]
  83. Tetreault, L.A.; Dettori, J.R.; Wilson, J.R.; Singh, A.; Nouri, A.; Fehlings, M.G.; Brodt, E.D.; Jacobs, W.B. Systematic review of magnetic resonance imaging characteristics that affect treatment decision making and predict clinical outcome in patients with cervical spondylotic myelopathy. Spine 2013, 38, S89–S110. [Google Scholar] [CrossRef]
  84. Taylor, A. Mechanism and treatment of spinal-cord disorders associated with cervical spondylosis. Lancet 1953, 261, 717–720. [Google Scholar] [CrossRef]
  85. Xing, R.; Zhou, G.; Chen, Q.; Liang, Y.; Dong, J. MRI to measure cervical sagittal parameters: A comparison with plain radiographs. Arch. Orthop. Trauma Surg. 2017, 137, 451–455. [Google Scholar] [CrossRef] [PubMed]
  86. Scheer, J.K.; Tang, J.A.; Smith, J.S.; Acosta, F.L.; Protopsaltis, T.S.; Blondel, B.; Bess, S.; Shaffrey, C.I.; Deviren, V.; Lafage, V. Cervical spine alignment, sagittal deformity, and clinical implications: A review. J. Neurosurg. Spine 2013, 19, 141–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Waly, F.J.; Abduljabbar, F.H.; Fortin, M.; Nooh, A.; Weber, M. Preoperative computed tomography myelography parameters as predictors of outcome in patients with degenerative cervical myelopathy: Results of a systematic review. Glob. Spine J. 2017, 7, 521–528. [Google Scholar] [CrossRef]
  88. Naderi, S.; Özgen, S.; Pamir, M.N.; Özek, M.M.; Erzen, C. Cervical spondylotic myelopathy: Surgical results and factors affecting prognosis. Neurosurgery 1998, 43, 43–49. [Google Scholar] [CrossRef] [PubMed]
  89. Nardone, R.; Höller, Y.; Brigo, F.; Frey, V.; Lochner, P.; Leis, S.; Golaszewski, S.; Trinka, E. The contribution of neurophysiology in the diagnosis and management of cervical spondylotic myelopathy: A review. Spinal Cord 2016, 54, 756–766. [Google Scholar] [CrossRef] [PubMed]
  90. Bischoff, C.; Meyer, B.U.; Machetanz, J.; Conrad, B. The value of magnetic stimulation in the diagnosis of radiculopathies. Muscle Nerve: Off. J. Am. Assoc. Electrodiagn. Med. 1993, 16, 154–161. [Google Scholar] [CrossRef] [PubMed]
  91. Bednařík, J.; Kadaňka, Z.; Voháňka, S.; Novotný, O.; Šurelová, D.; Filipovičová, D.; Prokeš, B. The value of somatosensory and motor evoked potentials in pre-clinical spondylotic cervical cord compression. Eur. Spine J. 1998, 7, 493–500. [Google Scholar] [CrossRef] [PubMed]
  92. Bednařík, J.; Sládková, D.; Kadaňka, Z.; Dušek, L.; Keřkovský, M.; Voháňka, S.; Novotný, O.; Urbánek, I.; Němec, M. Are subjects with spondylotic cervical cord encroachment at increased risk of cervical spinal cord injury after minor trauma? J. Neurol. Neurosurg. Psychiatry 2011, 82, 779–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bednarik, J.; Kadanka, Z.; Dusek, L.; Kerkovsky, M.; Vohanka, S.; Novotny, O.; Urbanek, I.; Kratochvilova, D. Presymptomatic spondylotic cervical myelopathy: An updated predictive model. Eur. Spine J. 2008, 17, 421–431. [Google Scholar] [CrossRef] [Green Version]
  94. Wilson, J.R.; Barry, S.; Fischer, D.J.; Skelly, A.C.; Arnold, P.M.; Riew, K.D.; Shaffrey, C.I.; Traynelis, V.C.; Fehlings, M.G. Frequency, timing, and predictors of neurological dysfunction in the nonmyelopathic patient with cervical spinal cord compression, canal stenosis, and/or ossification of the posterior longitudinal ligament. Spine 2013, 38, S37–S54. [Google Scholar] [CrossRef]
  95. Feng, X.; Hu, Y.; Ma, X. Progression Prediction of Mild Cervical Spondylotic Myelopathy by Somatosensory-evoked Potentials. Spine 2020, 45, E560–E567. [Google Scholar] [CrossRef] [PubMed]
  96. Dvorak, J.; Sutter, M.; Herdmann, J. Cervical myelopathy: Clinical and neurophysiological evaluation. Aging Spine 2005, 12, 99–105. [Google Scholar]
  97. Tsiptsios, I.; Fotiou, F.; Sitzoglou, K.; Fountoulakis, K. Neurophysiological investigation of cervical spondylosis. Electromyogr. Clin. Neurophysiol. 2001, 41, 305–313. [Google Scholar] [PubMed]
  98. Liu, H.; MacMillian, E.L.; Jutzeler, C.R.; Ljungberg, E.; MacKay, A.L.; Kolind, S.H.; Mädler, B.; Li, D.K.; Dvorak, M.F.; Curt, A. Assessing structure and function of myelin in cervical spondylotic myelopathy: Evidence of demyelination. Neurology 2017, 89, 602–610. [Google Scholar] [CrossRef] [PubMed]
  99. Capone, F.; Tamburelli, F.C.; Pilato, F.; Profice, P.; Ranieri, F.; Di Iorio, R.; Iodice, F.; Musumeci, G.; Di Lazzaro, V. The role of motor-evoked potentials in the management of cervical spondylotic myelopathy. Spine J. 2013, 13, 1077–1079. [Google Scholar] [CrossRef] [PubMed]
  100. Michaud, J. Chapter 11—Peripheral Nerves. In Essential Applications of Musculoskeletal Ultrasound in Rheumatology; Wakefield, R.J., D’Agostino, M.A., Eds.; W.B. Saunders: Philadelphia, PA, USA, 2010; pp. 121–136. [Google Scholar]
  101. Mowforth, O.D.; Davies, B.M.; Goh, S.; O’Neill, C.P.; Kotter, M.R. Research inefficiency in degenerative cervical myelopathy: Findings of a systematic review on research activity over the past 20 years. Glob. Spine J. 2020, 10, 476–485. [Google Scholar] [CrossRef] [Green Version]
  102. Ellingson, B.M.; Cohen-Adad, J. Chapter 3.1—Diffusion-Weighted Imaging of the Spinal Cord. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 123–145. [Google Scholar]
  103. Grabher, P.; Mohammadi, S.; Trachsler, A.; Friedl, S.; David, G.; Sutter, R.; Weiskopf, N.; Thompson, A.J.; Curt, A.; Freund, P. Voxel-based analysis of grey and white matter degeneration in cervical spondylotic myelopathy. Sci. Rep. 2016, 6, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Grabher, P.; Mohammadi, S.; David, G.; Freund, P. Neurodegeneration in the Spinal Ventral Horn Prior to Motor Impairment in Cervical Spondylotic Myelopathy. J. Neurotrauma 2017, 34, 2329–2334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Martin, A.R.; De Leener, B.; Cohen-Adad, J.; Cadotte, D.W.; Nouri, A.; Wilson, J.R.; Tetreault, L.; Crawley, A.P.; Mikulis, D.J.; Ginsberg, H. Can microstructural MRI detect subclinical tissue injury in subjects with asymptomatic cervical spinal cord compression? A prospective cohort study. BMJ Open 2018, 8, e019809. [Google Scholar] [CrossRef]
  106. Yoo, W.-K.; Kim, T.H.; Hai, D.M.; Sundaram, S.; Yang, Y.M.; Park, M.S.; Kim, Y.C.; Kwak, Y.H.; Ohn, S.H.; Kim, S.W. Correlation of magnetic resonance diffusion tensor imaging and clinical findings of cervical myelopathy. Spine J. 2013, 13, 867–876. [Google Scholar] [CrossRef] [PubMed]
  107. Martin, A.R.; De Leener, B.; Cohen-Adad, J.; Kalsi-Ryan, S.; Cadotte, D.W.; Wilson, J.R.; Tetreault, L.; Nouri, A.; Crawley, A.; Mikulis, D.J. Monitoring for myelopathic progression with multiparametric quantitative MRI. PLoS ONE 2018, 13, e0195733. [Google Scholar]
  108. Wang, K.; Chen, Z.; Zhang, F.; Song, Q.; Hou, C.; Tang, Y.; Wang, J.; Chen, S.; Bian, Y.; Hao, Q. Evaluation of DTI parameter ratios and diffusion tensor tractography grading in the diagnosis and prognosis prediction of cervical spondylotic myelopathy. Spine 2017, 42, E202–E210. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, H.; Guan, L.; Hai, Y.; Liu, Y.; Ding, H.; Chen, X. Multi-shot echo-planar diffusion tensor imaging in cervical spondylotic myelopathy: A longitudinal study. Bone Jt. J. 2020, 102, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  110. Martin, A.R.; Aleksanderek, I.; Cohen-Adad, J.; Tarmohamed, Z.; Tetreault, L.; Smith, N.; Cadotte, D.W.; Crawley, A.; Ginsberg, H.; Mikulis, D.J. Translating state-of-the-art spinal cord MRI techniques to clinical use: A systematic review of clinical studies utilizing DTI, MT, MWF, MRS, and fMRI. NeuroImage Clin. 2016, 10, 192–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Martin, A.; De Leener, B.; Cohen-Adad, J.; Cadotte, D.; Kalsi-Ryan, S.; Lange, S.; Tetreault, L.; Nouri, A.; Crawley, A.; Mikulis, D. Clinically feasible microstructural MRI to quantify cervical spinal cord tissue injury using DTI, MT, and T2*-weighted imaging: Assessment of normative data and reliability. Am. J. Neuroradiol. 2017, 38, 1257–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Summers, P.E.; Brooks, J.C.W.; Cohen-Adad, J. Chapter 4.1—Spinal Cord fMRI. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 221–239. [Google Scholar]
  113. Solanky, B.S.; De Vita, E. Chapter 5.1—Single Voxel MR Spectroscopy in the Spinal Cord: Technical Challenges and Clinical Applications. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 267–290. [Google Scholar]
  114. Laule, C.; MacKay, A. Chapter 3.5—T2 Relaxation. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 181–206. [Google Scholar]
  115. Kim, M.; Cercignani, M. Chapter 3.4—Magnetization Transfer. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 164–180. [Google Scholar]
  116. Brooks, J.C.W. Chapter 4.2—Physiological Noise Modeling and Analysis for Spinal Cord fMRI. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 240–257. [Google Scholar]
  117. Assaf, Y.; Alexander, D.C. Chapter 3.3—Advanced Methods to Study White Matter Microstructure. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 156–163. [Google Scholar]
  118. Abdel-Aziz, K.; Ciccarelli, O. Chapter 1.1—Rationale for Quantitative MRI of the Human Spinal Cord and Clinical Applications. In Quantitative MRI of the Spinal Cord; Cohen-Adad, J., Wheeler-Kingshott, C.A.M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 3–21. [Google Scholar]
  119. Chavhan, G.B.; Babyn, P.S.; Thomas, B.; Shroff, M.M.; Haacke, E.M. Principles, techniques, and applications of T2 *-based MR imaging and its special applications. Radiographics 2009, 29, 1433–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Battiston, M.; Schneider, T.; Prados, F.; Grussu, F.; Yiannakas, M.C.; Ourselin, S.; Gandini Wheeler-Kingshott, C.A.; Samson, R.S. Fast and reproducible in vivo T1 mapping of the human cervical spinal cord. Magn. Reson. Med. 2018, 79, 2142–2148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.; Horsley, T.; Weeks, L. PRISMA extension for scoping reviews (PRISMA-ScR): Checklist and explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Albistegui-Dubois, R.; Marehbian, J.; Newton, J.M.; Dong, Y.; Holly, L.T.; Yan, X.; Dobkin, B.H. Compensatory cerebral adaptations before and evolving changes after surgical decompression in cervical spondylotic myelopathy: Laboratory investigation. J. Neurosurg. Spine 2008, 9, 538–551. [Google Scholar]
  123. Aleksanderek, I.K.; Stevens, T.; Goncalves, S.; Bartha, R.; Duggal, N. Investigating metabolic and functional profiles of mild and moderate cervical spondylotic myelopathy: A MRS and fMRI study. Spine J. 2015, 15, S201. [Google Scholar] [CrossRef]
  124. Banaszek, A.; Bladowska, J.; Szewczyk, P.; Podgorski, P.; Sasiadek, M. Usefulness of diffusion tensor MR imaging in the assessment of intramedullary changes of the cervical spinal cord in different stages of degenerative spine disease. Eur. Spine J. 2014, 23, 1523–1530. [Google Scholar] [CrossRef] [Green Version]
  125. Baucher, G.; Rasoanandrianina, H.; Levy, S.; Pini, L.; Troude, L.; Roche, P.H.; Callot, V. T1 Mapping for Microstructural Assessment of the Cervical Spinal Cord in the Evaluation of Patients with Degenerative Cervical Myelopathy. AJNR. Am. J. Neuroradiol. 2021, 42, 1348–1357. [Google Scholar] [CrossRef] [PubMed]
  126. Bhagavatula, I.D.; Shukla, D.; Sadashiva, N.; Saligoudar, P.; Prasad, C.; Bhat, D.I. Functional cortical reorganization in cases of cervical spondylotic myelopathy and changes associated with surgery. Neurosurg. Focus 2016, 40, E2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Bhosale, S.; Ingale, P.; Srivastava, S.; Marathe, N.; Bhide, P. Diffusion tensor imaging as an additional postoperative prognostic predictor factor in cervical myelopathy patients: An observational study. J. Craniovertebral Junction Spine 2019, 10, 10–13. [Google Scholar]
  128. Chen, X.; Kong, C.; Feng, S.; Guan, H.; Yu, Z.; Cui, L.; Wang, Y. Magnetic resonance diffusion tensor imaging of cervical spinal cord and lumbosacral enlargement in patients with cervical spondylotic myelopathy. J. Magn. Reson. Imaging 2016, 43, 1484–1491. [Google Scholar] [CrossRef] [PubMed]
  129. Chen, Z.; Zhao, R.; Wang, Q.; Yu, C.; Li, F.; Liang, M.; Zong, Y.; Zhao, Y.; Xiong, W.; Su, Z.; et al. Functional Connectivity Changes of the Visual Cortex in the Cervical Spondylotic Myelopathy Patients: A Resting-State fMRI Study. Spine 2020, 45, E272–E279. [Google Scholar] [CrossRef] [PubMed]
  130. Cloney, M.B.; Smith, Z.A.; Weber, K.A.; Parrish, T.B. Quantitative Magnetization Transfer MRI Measurements of the Anterior Spinal Cord Region are Associated with Clinical Outcomes in Cervical Spondylotic Myelopathy. Spine 2018, 43, 675–680. [Google Scholar] [CrossRef]
  131. Cui, J.-L.; Li, X.; Chan, T.-Y.; Mak, K.-C.; Luk, K.D.-K.; Hu, Y. Quantitative assessment of column-specific degeneration in cervical spondylotic myelopathy based on diffusion tensor tractography. Eur. Spine J. 2015, 24, 41–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Cui, L.; Chen, X.; Liu, Y.; Zhang, Y.; Kong, C.; Guan, Y. Changes in diffusion tensor imaging indices of the lumbosacral enlargement correlate with cervical spinal cord changes and clinical assessment in patients with cervical spondylotic myelopathy. Clin. Neurol. Neurosurg. 2019, 186, 105282. [Google Scholar] [CrossRef] [PubMed]
  133. Duggal, N.; Rabin, D.; Bartha, R.; Barry, R.L.; Gati, J.S.; Kowalczyk, I.; Fink, M. Brain reorganization in patients with spinal cord compression evaluated using fMRI. Neurology 2010, 74, 1048–1054. [Google Scholar] [CrossRef] [PubMed]
  134. Ellingson, B.M.; Salamon, N.; Grinstead, J.W.; Holly, L.T. Diffusion tensor imaging predicts functional impairment in mild-to-moderate cervical spondylotic myelopathy. Spine J. Off. J. North Am. Spine Soc. 2014, 14, 2589–2597. [Google Scholar] [CrossRef] [Green Version]
  135. Ellingson, B.M.; Salamon, N.; Hardy, A.J.; Holly, L.T. Prediction of Neurological Impairment in Cervical Spondylotic Myelopathy using a Combination of Diffusion MRI and Proton MR Spectroscopy. PLoS ONE 2015, 10, e0139451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Guo, X.; Yang, X.; Chen, X.; Zhao, R.; Song, Y.; Liang, M.; Sun, H.; Xue, Y. Enhanced Information Flow From Cerebellum to Secondary Visual Cortices Leads to Better Surgery Outcome in Degenerative Cervical Myelopathy Patients: A Stochastic Dynamic Causal Modeling Study With Functional Magnetic Resonance Imaging. Front. Hum. Neurosci. 2021, 15, 632829. [Google Scholar]
  137. Guo, X.; Yang, X.; Chen, X.; Zhao, R.; Song, Y.; Liang, M.; Sun, H.; Xue, Y. The Evaluation and Prediction of Laminoplasty Surgery Outcome in Patients with Degenerative Cervical Myelopathy Using Diffusion Tensor MRI. AJNR. Am. J. Neuroradiol. 2020, 41, 1745–1753. [Google Scholar]
  138. Hassan, T.A.A.E.H.; Assad, R.E.; Belal, S.A. MR diffusion tensor imaging of the spinal cord: Can it help in early detection of cervical spondylotic myelopathy and assessment of its severity? Egypt. J. Radiol. Nucl. Med. 2019, 50, 62. [Google Scholar] [CrossRef] [Green Version]
  139. He, Z.; Wang, N.; Kang, L.; Cui, J.; Wan, Y. Analysis of pathological parameters of cervical spondylotic myelopathy using magnetic resonance imaging. Clin. Neurol. Neurosurg. 2020, 189, 105631. [Google Scholar] [CrossRef] [PubMed]
  140. Holly, L.T.; Wang, C.; Salamon, N.; Woodworth, D.C.; Ellingson, B.M. Neck disability in patients with cervical spondylosis is associated with altered brain functional connectivity. J. Clin. Neurosci. 2019, 69, 149–154. [Google Scholar] [CrossRef] [PubMed]
  141. Holly, L.T.; Wang, C.; Salamon, N.; Woodworth, D.C.; Ellingson, B.M. New diffusion metrics for spondylotic myelopathy at an early clinical stage. Eur. Radiol. 2012, 22, 1797–1802. [Google Scholar]
  142. Iwasaki, M.; Yokohama, T.; Oura, D.; Furuya, S.; Niiya, Y.; Okuaki, T. Decreased Value of Highly Accurate Fractional Anisotropy Using 3-Tesla ZOOM Diffusion Tensor Imaging After Decompressive Surgery in Patients with Cervical Spondylotic Myelopathy: Aligned Fibers Effect. World Neurosurg. X 2019, 4, 100056. [Google Scholar] [CrossRef]
  143. Jurova, B.; Mechl, M.; Kerkovsky, M.; Sprlakova-Pukova, A.; Kadanka, Z.; Nemec, M.; Bednarik, J.; Kovalova, I.; Dusek, L. Spinal Cord MR Diffusion Properties in Patients with Degenerative Cervical Cord Compression. J. Neuroimaging 2017, 27, 149–157. [Google Scholar]
  144. Kanchiku, T.; Imajo, Y.; Suzuki, H.; Yoshida, Y.; Nishida, N.; Taguchi, T.; Suetomi, Y.; Nishijima, S. Application of diffusion tensor imaging for the diagnosis of segmental level of dysfunction in cervical spondylotic myelopathy. Spinal Cord 2016, 54, 390–395. [Google Scholar]
  145. Kara, B.; Celik, A.; Karadereler, S.; Ulusoy, L.; Ganiyusufoglu, K.; Onat, L.; Mutlu, A.; Ornek, I.; Sirvanci, M.; Hamzaoglu, A. The role of DTI in early detection of cervical spondylotic myelopathy: A preliminary study with 3-T MRI. Neuroradiology 2011, 53, 609–616. [Google Scholar] [CrossRef] [PubMed]
  146. Kerkovsky, M.; Jakubcova, B.; Mechl, M.; Kadanka, Z.; Kadanka Jr, Z.; Nemec, M.; Kovalova, I.; Bednarik, J. Multifactorial determination of the spinal cord diffusion properties in patients with cervical spondylotic spinal cord compression: A diffusion tensor imaging study. Neuroradiology 2015, 57, S133. [Google Scholar]
  147. Kowalczyk, I.; Bartha, R.; Duggal, N. Proton magnetic resonance spectroscopy of the motor cortex in cervical myelopathy. Brain 2012, 135, 461–468. [Google Scholar] [CrossRef] [Green Version]
  148. Kowalczyk, I.; Bartha, R.; Duggal, N. Proton magnetic resonance spectroscopy of the motor cortex in cervical spondylotic myelopathy. Can. J. Neurol. Sci. 2010, 37, S30. [Google Scholar]
  149. Lee, S.; Chung, T.-S.; Kim, S.; Yoo, Y.H.; Yoon, C.-S.; Lee, Y.H.; Suh, J.-S.; Jeong, E.-K.; Kim, I.S.; Park, J.H. Accuracy of diffusion tensor imaging for diagnosing cervical spondylotic myelopathy in patients showing spinal cord compression. Korean J. Radiol. 2015, 16, 1303–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Liu, X.; Qian, W.; Jin, R.; Li, X.; Luk, K.D.; Wu, E.X.; Hu, Y. Amplitude of Low Frequency Fluctuation (ALFF) in the Cervical Spinal Cord with Stenosis: A Resting State fMRI Study. PLoS ONE 2016, 11, e0167279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Maier, I.L.; Hofer, S.; Eggert, E.; Schregel, K.; Psychogios, M.-N.; Frahm, J.; Bähr, M.; Liman, J. T1 Mapping Quantifies Spinal Cord Compression in Patients with Various Degrees of Cervical Spinal Canal Stenosis. Front. Neurol. 2020, 11, 574604. [Google Scholar] [CrossRef]
  152. Maki, S.; Koda, M.; Kitamura, M.; Inada, T.; Kamiya, K.; Ota, M.; Iijima, Y.; Saito, J.; Masuda, Y.; Matsumoto, K.; et al. Diffusion tensor imaging can predict surgical outcomes of patients with cervical compression myelopathy. Eur. Spine J. 2017, 26, 2459–2466. [Google Scholar] [CrossRef]
  153. Maki, S.; Koda, M.; Ota, M.; Oikawa, Y.; Kamiya, K.; Inada, T.; Furuya, T.; Takahashi, K.; Masuda, Y.; Matsumoto, K.; et al. Reduced Field-of-View Diffusion Tensor Imaging of the Spinal Cord Shows Motor Dysfunction of the Lower Extremities in Patients with Cervical Compression Myelopathy. Spine 2018, 43, 89–96. [Google Scholar] [CrossRef]
  154. Mamata, H.; Jolesz, F.A.; Maier, S.E. Apparent diffusion coefficient and fractional anisotropy in spinal cord: Age and cervical spondylosis-related changes. J. Magn. Reson. Imaging 2005, 22, 38–43. [Google Scholar] [CrossRef] [PubMed]
  155. Murphy, R.K.; Sun, P.; Han, R.H.; Griffin, K.J.; Wagner, J.; Yarbrough, C.K.; Wright, N.M.; Dorward, I.G.; Riew, K.D.; Kelly, M.P.; et al. Fractional anisotropy to quantify cervical spondylotic myelopathy severity. J. Neurosurg. Sci. 2018, 62, 406–412. [Google Scholar] [CrossRef] [PubMed]
  156. Nagashima, H.; Nanjo, Y.; Teshima, R.; Morio, Y.; Meshitsuka, S.; Yamane, K. High-resolution nuclear magnetic resonance spectroscopic study of metabolites in the cerebrospinal fluid of patients with cervical myelopathy and lumbar radiculopathy. Eur. Spine J. 2010, 19, 1363–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Nischal, N.; Tripathi, S.; Singh, J.P. Quantitative Evaluation of the Diffusion Tensor Imaging Matrix Parameters and the Subsequent Correlation with the Clinical Assessment of Disease Severity in Cervical Spondylotic Myelopathy. Asian Spine J. 2020, 15, 808–816. [Google Scholar] [CrossRef]
  158. Nukala, M.; Abraham, J.; Khandige, G.; Shetty, B.K.; Rao, A.p.a. Efficacy of diffusion tensor imaging in identification of degenerative cervical spondylotic myelopathy. Eur. J. Radiol. Open 2019, 6, 16–23. [Google Scholar] [CrossRef] [Green Version]
  159. Paliwal, M.; Smith, Z.A.; Weber, K.A.; Mackey, S.; Hopkins, B.S.; Dahdaleh, N.S.; Cantrell, D.R.; Parrish, T.D.; Hoggarth, M.A.; Elliott, J.M.; et al. Magnetization Transfer Ratio and Morphometrics of the Spinal Cord Associates with Surgical Recovery in Patients with Degenerative Cervical Myelopathy. World Neurosurg. 2020, 144, e939–e947. [Google Scholar] [CrossRef]
  160. Peng, X.; Tan, Y.; He, L.; Ou, Y. Alterations of functional connectivity between thalamus and cortex before and after decompression in cervical spondylotic myelopathy patients: A resting-state functional MRI study. NeuroReport 2020, 31, 365–371. [Google Scholar] [CrossRef]
  161. Rajasekaran, S.; Kanna, R.M.; Balamurali, G.; Shetty, A.P.; Yerramshetty, J.S.; Chittode, V.S. The assessment of neuronal status in normal and cervical spondylotic myelopathy using diffusion tensor imaging. Spine 2014, 39, 1183–1189. [Google Scholar] [CrossRef]
  162. Rajasekaran, S.; Kanna, R.M.; Chittode, V.S.; Maheswaran, A.; Aiyer, S.N.; Shetty, A.P. Efficacy of Diffusion Tensor Imaging Indices in Assessing Postoperative Neural Recovery in Cervical Spondylotic Myelopathy. Spine 2017, 42, 8–13. [Google Scholar] [CrossRef]
  163. Salamon, N.; Ellingson, B.M.; Nagarajan, R.; Gebara, N.; Thomas, A.; Holly, L.T. Proton magnetic resonance spectroscopy of human cervical spondylosis at 3T. Spinal Cord 2013, 51, 558–563. [Google Scholar] [CrossRef] [Green Version]
  164. Salamon, N.; Woodworth, D.C.; Holly, L.T.; Ellingson, B.M. Resting-State Functional Magnetic Resonance Imaging Connectivity of the Brain Is Associated with Altered Sensorimotor Function in Patients with Cervical Spondylosis. World Neurosurg. 2018, 119, e740–e749. [Google Scholar]
  165. Sato, T.; Horikoshi, T.; Watanabe, A.; Uchida, M.; Ishigame, K.; Araki, T.; Kinouchi, H. Evaluation of cervical myelopathy using apparent diffusion coefficient measured by diffusion-weighted imaging. AJNR. Am. J. Neuroradiol. 2012, 33, 388–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Severino, R.; Nouri, A.; Tessitore, E. Degenerative cervical myelopathy: How to identify the best responders to surgery? J. Clin. Med. 2020, 9, 759. [Google Scholar] [CrossRef] [PubMed]
  167. Shabani, S.; Kaushal, M.; Budde, M.; Schmit, B.; Wang, M.C.; Kurpad, S. Comparison between quantitative measurements of diffusion tensor imaging and T2 signal intensity in a large series of cervical spondylotic myelopathy patients for assessment of disease severity and prognostication of recovery. J. Neurosurg. Spine 2019, 31, 473–479. [Google Scholar] [CrossRef] [PubMed]
  168. Song, T.; Chen, W.-J.; Huang, J.-W.; Cai, M.-J.; Dong, T.-F.; Li, T.-S.; Yang, B.; Zhao, H.-P. Diffusion tensor imaging in the cervical spinal cord. Eur. Spine J. 2011, 20, 422–428. [Google Scholar] [CrossRef] [Green Version]
  169. Su, Q.; Zhao, R.; Guo, X.; Wang, S.; Tu, H.; Yang, F. Identification and Therapeutic Outcome Prediction of Cervical Spondylotic Myelopathy Based on the Functional Connectivity From Resting-State Functional MRI Data: A Preliminary Machine Learning Study. Front. Neurol. 2021, 12, 711880. [Google Scholar] [CrossRef]
  170. Suleiman, L.I. High-resolution magnetization transfer MRI in patients with cervical spondylotic myelopathy. J. Clin. Neurosci. 2018, 51, 57–61. [Google Scholar] [CrossRef]
  171. Taha Ali, T.F.; Badawy, A.E. Feasibility of 1H-MR Spectroscopy in evaluation of cervical spondylotic myelopathy. Egypt. J. Radiol. Nucl. Med. 2013, 44, 93–99. [Google Scholar] [CrossRef] [Green Version]
  172. Takenaka, S.; Kan, S.; Seymour, B.; Makino, T.; Sakai, Y.; Kushioka, J.; Tanaka, H.; Watanabe, Y.; Shibata, M.; Yoshikawa, H.; et al. Resting-state Amplitude of Low-frequency Fluctuation is a Potentially Useful Prognostic Functional Biomarker in Cervical Myelopathy. Clin. Orthop. Relat. Res. 2020, 478, 1667–1680. [Google Scholar] [CrossRef]
  173. Takenaka, S.; Kan, S.; Seymour, B.; Makino, T.; Sakai, Y.; Kushioka, J.; Tanaka, H.; Watanabe, Y.; Shibata, M.; Yoshikawa, H.; et al. Towards prognostic functional brain biomarkers for cervical myelopathy: A resting-state fMRI study. Sci. Rep. 2019, 9, 10456. [Google Scholar] [CrossRef] [Green Version]
  174. Tan, Y.; Zhou, F.; Liu, Z.; Wu, L.; Zeng, X.; Gong, H.; He, L. Alteration of cerebral regional homogeneity within sensorimotor network in patients with cervical spondylotic myelopathy after spinal cord decompression: A resting-state functional MRI study. Chin. J. Radiol. 2016, 50, 495–499. [Google Scholar]
  175. Tian, X.; Zhang, L.; Zhang, X.; Meng, L.; Li, X. Correlations between preoperative diffusion tensor imaging and surgical outcome in patients with cervical spondylotic myelopathy. Am. J. Transl. Res. 2021, 13, 11461–11471. [Google Scholar] [PubMed]
  176. Toktas, Z.O.; Kilic, T.; Konya, D.; Tanrikulu, B.; Koban, O. Diffusion tensor imaging of cervical spinal cord: A quantitative diagnostic tool in cervical spondylotic myelopathy. J. Craniovertebral Junction Spine 2016, 7, 26–30. [Google Scholar] [CrossRef] [PubMed]
  177. Uda, T.; Takami, T.; Tsuyuguchi, N.; Sakamoto, S.; Yamagata, T.; Ikeda, H.; Nagata, T.; Ohata, K. Assessment of cervical spondylotic myelopathy using diffusion tensor magnetic resonance imaging parameter at 3.0 tesla. Spine 2013, 38, 407–414. [Google Scholar] [CrossRef]
  178. Ulubaba, H.E.; Saglik, S.; Yildirim, I.O.; Durak, M.A. Effectiveness of Diffusion Tensor Imaging in Determining Cervical Spondylotic Myelopathy. Turk. Neurosurg. 2021, 31, 67–72. [Google Scholar] [CrossRef]
  179. Vedantam, A.; Rao, A.; Kurpad, S.N.; Jirjis, M.B.; Eckardt, G.; Schmit, B.D.; Wang, M.C. Diffusion Tensor Imaging Correlates with Short-Term Myelopathy Outcome in Patients with Cervical Spondylotic Myelopathy. World Neurosurg. 2017, 97, 489–494. [Google Scholar] [CrossRef] [Green Version]
  180. Wang, C.; Salamon, N.; Laiwalla, A.; Holly, L.T.; Ellingson, B.M.; Islam, S. Supraspinal functional and structural plasticity in patients undergoing surgery for degenerative cervical myelopathy. J. Neurosurg. Spine 2021, 35, 185–193. [Google Scholar] [CrossRef]
  181. Wang, K.Y.; Idowu, O.; Orman, G.; Izbudak, I.; Thompson, C.B.; Myers, C.; Riley, L.H.; Carrino, J.A.; Flammang, A.; Gilson, W.; et al. Tract-Specific Diffusion Tensor Imaging in Cervical Spondylotic Myelopathy Before and After Decompressive Spinal Surgery: Preliminary Results. Clin. Neuroradiol. 2017, 27, 61–69. [Google Scholar] [CrossRef]
  182. Wen, C.Y.; Cui, J.L.; Liu, H.S.; Mak, K.C.; Cheung, W.Y.; Luk, K.D.K.; Hu, Y. Is diffusion anisotropy a biomarker for disease severity and surgical prognosis of cervical spondylotic myelopathy. Radiology 2014, 270, 197–204. [Google Scholar] [CrossRef]
  183. Xiangshui, M.; Xiangjun, C.; Xiaoming, Z.; Qingshi, Z.; Yi, C.; Chuanqiang, Q.; Xiangxing, M.; Chuanfu, L.; Jinwen, H. 3 T magnetic resonance diffusion tensor imaging and fibre tracking in cervical myelopathy. Clin. Radiol. 2010, 65, 465–473. [Google Scholar] [CrossRef]
  184. Zhang, M.-Z.; Liu, J.-F.; Jin, D.; Wang, C.-J.; Zhao, Q.; Lang, N.; Yuan, H.-S.; Ou-Yang, H.-Q.; Liu, X.-G.; Liu, Z.-J.; et al. Utility of Advanced DWI in the Detection of Spinal Cord Microstructural Alterations and Assessment of Neurologic Function in Cervical Spondylotic Myelopathy Patients. J. Magn. Reson. Imaging 2021, 55, 930–940. [Google Scholar] [CrossRef]
  185. Zheng, W.; Chen, H.; Wang, N.; Jiang, X.; Liang, Y.; Xiao, W.; Zhong, B.; Ju, H.; Luo, J.; Wen, S.; et al. Application of Diffusion Tensor Imaging Cutoff Value to Evaluate the Severity and Postoperative Neurologic Recovery of Cervical Spondylotic Myelopathy. World Neurosurg. 2018, 118, e849–e855. [Google Scholar] [CrossRef]
  186. Taylor, A.J.; Salerno, M.; Dharmakumar, R.; Jerosch-Herold, M. T1 mapping: Basic techniques and clinical applications. JACC Cardiovasc. Imaging 2016, 9, 67–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Watanabe, A.; Benneker, L.M.; Boesch, C.; Watanabe, T.; Obata, T.; Anderson, S.E. Classification of intervertebral disk degeneration with axial T2 mapping. Am. J. Roentgenol. 2007, 189, 936–942. [Google Scholar] [CrossRef] [PubMed]
  188. Deoni, S.C.; Rutt, B.K.; Peters, T.M. Rapid combined T1 and T2 mapping using gradient recalled acquisition in the steady state. Magn. Reson. Med. 2003, 49, 515–526. [Google Scholar] [CrossRef]
  189. Henderson, E.; McKinnon, G.; Lee, T.-Y.; Rutt, B.K. A fast 3D look-locker method for volumetric T1 mapping. Magn. Reson. Imaging 1999, 17, 1163–1171. [Google Scholar] [CrossRef]
  190. Wang, X.; Joseph, A.A.; Kalentev, O.; Merboldt, K.-D.; Voit, D.; Roeloffs, V.B.; van Zalk, M.; Frahm, J. High-resolution myocardial T 1 mapping using single-shot inversion recovery fast low-angle shot MRI with radial undersampling and iterative reconstruction. Br. J. Radiol. 2016, 89, 20160255. [Google Scholar] [CrossRef]
  191. Nöth, U.; Shrestha, M.; Schüre, J.-R.; Deichmann, R. Quantitative in vivo T2 mapping using fast spin echo techniques–A linear correction procedure. Neuroimage 2017, 157, 476–485. [Google Scholar] [CrossRef] [PubMed]
  192. Lommers, E.; Simon, J.; Reuter, G.; Delrue, G.; Dive, D.; Degueldre, C.; Balteau, E.; Phillips, C.; Maquet, P. Multiparameter MRI quantification of microstructural tissue alterations in multiple sclerosis. NeuroImage Clin. 2019, 23, 101879. [Google Scholar] [CrossRef]
  193. Steenwijk, M.D.; Vrenken, H.; Jonkman, L.E.; Daams, M.; Geurts, J.J.; Barkhof, F.; Pouwels, P.J. High-resolution T1-relaxation time mapping displays subtle, clinically relevant, gray matter damage in long-standing multiple sclerosis. Mult. Scler. J. 2016, 22, 1279–1288. [Google Scholar] [CrossRef]
  194. Rasoanandrianina, H. Regional T1 mapping of the whole cervical spinal cord using an optimized MP2RAGE sequence. NMR Biomed. 2019, 32, e4142. [Google Scholar] [CrossRef] [PubMed]
  195. Marinelli, N.; Haughton, V.M.; Anderson, P. T2 relaxation times correlated with stage of lumbar intervertebral disk degeneration and patient age. Am. J. Neuroradiol. 2010, 31, 1278–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Pachowsky, M.L.; Kleyer, A.; Wegener, L.; Langenbach, A.; Simon, D.; Janka, R.; May, M.; Welsch, G.H. Quantitative T2 mapping shows increased degeneration in adjacent intervertebral discs following kyphoplasty. Cartilage 2020, 11, 152–159. [Google Scholar] [CrossRef] [PubMed]
  197. Raudner, M.; Schreiner, M.M.; Hilbert, T.; Kober, T.; Weber, M.; Szelényi, A.; Windhager, R.; Juras, V.; Trattnig, S. Clinical implementation of accelerated T 2 mapping: Quantitative magnetic resonance imaging as a biomarker for annular tear and lumbar disc herniation. Eur. Radiol. 2021, 31, 3590–3599. [Google Scholar] [CrossRef] [PubMed]
  198. Chagawa, K.; Nishijima, S.; Kanchiku, T.; Imajo, Y.; Suzuki, H.; Yoshida, Y.; Taguchi, T. Normal values of diffusion tensor magnetic resonance imaging parameters in the cervical spinal cord. Asian Spine J. 2015, 9, 541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Dong, F.; Wu, Y.; Song, P.; Qian, Y.; Wang, Y.; Xu, L.; Yin, M.; Zhang, R.; Tao, H.; Ge, P. A preliminary study of 3.0-T magnetic resonance diffusion tensor imaging in cervical spondylotic myelopathy. Eur. Spine J. 2018, 27, 1839–1845. [Google Scholar] [CrossRef]
  200. Guan, X.; Fan, G.; Wu, X.; Gu, G.; Gu, X.; Zhang, H.; He, S. Diffusion tensor imaging studies of cervical spondylotic myelopathy: A systemic review and meta-analysis. PLoS ONE 2015, 10, e0117707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. d’Avanzo, S.; Ciavarro, M.; Pavone, L.; Pasqua, G.; Ricciardi, F.; Bartolo, M.; Solari, D.; Somma, T.; de Divitiis, O.; Cappabianca, P.; et al. The Functional Relevance of Diffusion Tensor Imaging in Patients with Degenerative Cervical Myelopathy. J. Clin. Med. 2020, 9, 1828. [Google Scholar] [CrossRef] [PubMed]
  202. Ibrahim, H.I.A.; Saleh, A. Role of DTI in cases of cervical spondylosis presented with compression myelopathy: Could it explain the clinical radiological mismatch?! Egypt. J. Radiol. Nucl. Med. 2018, 49, 441–446. [Google Scholar] [CrossRef]
  203. Jones, J.G.A.; Cen, S.Y.; Lebel, R.M.; Hsieh, P.C.; Law, M. Diffusion Tensor Imaging Correlates with the Clinical Assessment of Disease Severity in Cervical Spondylotic Myelopathy and Predicts Outcome following Surgery. Am. J. Neuroradiol. 2013, 34, 471–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Logothetis, N.K.; Pfeuffer, J. On the nature of the BOLD fMRI contrast mechanism. Magn. Reson. Imaging 2004, 22, 1517–1531. [Google Scholar] [CrossRef] [PubMed]
  205. Gomez-Anson, B.; MacManus, D.; Parker, G.; Davie, C.; Barker, G.; Moseley, I.; McDonald, W.; Miller, D. In vivo 1 H-magnetic resonance spectroscopy of the spinal cord in humans. Neuroradiology 2000, 42, 515–517. [Google Scholar] [CrossRef] [PubMed]
  206. Oh, J.; Saidha, S.; Chen, M.; Smith, S.A.; Prince, J.; Jones, C.; Diener-West, M.; Van Zijl, P.C.; Reich, D.S.; Calabresi, P.A. Spinal cord quantitative MRI discriminates between disability levels in multiple sclerosis. Neurology 2013, 80, 540–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Petrella, J.R.; Grossman, R.I.; McGowan, J.C.; Campbell, G.; Cohen, J.A. Multiple sclerosis lesions: Relationship between MR enhancement pattern and magnetization transfer effect. Am. J. Neuroradiol. 1996, 17, 1041–1049. [Google Scholar] [PubMed]
  208. Serbruyns, L.; Leunissen, I.; van Ruitenbeek, P.; Pauwels, L.; Caeyenberghs, K.; Solesio-Jofre, E.; Geurts, M.; Cuypers, K.; Meesen, R.L.; Sunaert, S. Alterations in brain white matter contributing to age-related slowing of task switching performance: The role of radial diffusivity and magnetization transfer ratio. Hum. Brain Mapp. 2016, 37, 4084–4098. [Google Scholar] [CrossRef] [PubMed]
  209. Hankins, J.S.; McCarville, M.B.; Loeffler, R.B.; Smeltzer, M.P.; Onciu, M.; Hoffer, F.A.; Li, C.-S.; Wang, W.C.; Ware, R.E.; Hillenbrand, C.M. R2 * magnetic resonance imaging of the liver in patients with iron overload. Blood J. Am. Soc. Hematol. 2009, 113, 4853–4855. [Google Scholar] [CrossRef] [PubMed]
  210. Rivera-Rivera, L.A.; Schubert, T.; Johnson, K.M. Measurements of cerebral blood volume using quantitative susceptibility mapping, R2 * relaxometry, and ferumoxytol-enhanced MRI. NMR Biomed. 2019, 32, e4175. [Google Scholar] [CrossRef] [PubMed]
  211. Núñez, M.T.; Hidalgo, C. Noxious Iron–calcium connections in Neurodegeneration. Front. Neurosci. 2019, 13, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Ward, R.J.; Zucca, F.A.; Duyn, J.H.; Crichton, R.R.; Zecca, L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014, 13, 1045–1060. [Google Scholar] [CrossRef] [Green Version]
  213. Crichton, R.; Ward, R. Metal-Based Neurodegeneration: From Molecular Mechanisms to Therapeutic Strategies; John Wiley & Sons: Hoboken, NJ, USA, 2013. [Google Scholar]
  214. Crichton, R.; Crichton, R.R.; Boelaert, J.R. Inorganic Biochemistry of Iron Metabolism: From Molecular Mechanisms to Clinical Consequences; John Wiley & Sons: Hoboken, NJ, USA, 2001. [Google Scholar]
  215. Craelius, W.; Migdal, M.; Luessenhop, C.; Sugar, A.; Mihalakis, I. Iron deposits surrounding multiple sclerosis plaques. Arch. Pathol. Lab. Med. 1982, 106, 397–399. [Google Scholar] [PubMed]
  216. Hametner, S.; Wimmer, I.; Haider, L.; Pfeifenbring, S.; Brück, W.; Lassmann, H. Iron and neurodegeneration in the multiple sclerosis brain. Ann. Neurol. 2013, 74, 848–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Haider, L.; Simeonidou, C.; Steinberger, G.; Hametner, S.; Grigoriadis, N.; Deretzi, G.; Kovacs, G.G.; Kutzelnigg, A.; Lassmann, H.; Frischer, J.M. Multiple sclerosis deep grey matter: The relation between demyelination, neurodegeneration, inflammation and iron. J. Neurol. Neurosurg. Psychiatry 2014, 85, 1386–1395. [Google Scholar] [CrossRef] [Green Version]
  218. Schuh, C.; Wimmer, I.; Hametner, S.; Haider, L.; Van Dam, A.-M.; Liblau, R.S.; Smith, K.J.; Probert, L.; Binder, C.J.; Bauer, J. Oxidative tissue injury in multiple sclerosis is only partly reflected in experimental disease models. Acta Neuropathol. 2014, 128, 247–266. [Google Scholar] [CrossRef] [Green Version]
  219. Bulk, M.; van der Weerd, L.; Breimer, W.; Lebedev, N.; Webb, A.; Goeman, J.J.; Ward, R.J.; Huber, M.; Oosterkamp, T.H.; Bossoni, L. Quantitative comparison of different iron forms in the temporal cortex of Alzheimer patients and control subjects. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef] [PubMed]
  220. Lee, J.-H.; Han, Y.-H.; Kang, B.-M.; Mun, C.-W.; Lee, S.-J.; Baik, S.-K. Quantitative assessment of subcortical atrophy and iron content in progressive supranuclear palsy and parkinsonian variant of multiple system atrophy. J. Neurol. 2013, 260, 2094–2101. [Google Scholar] [CrossRef] [PubMed]
  221. Swaiman, K.F. Hallervorden-Spatz syndrome and brain iron metabolism. Arch. Neurol. 1991, 48, 1285–1293. [Google Scholar] [CrossRef] [PubMed]
  222. Wiethoff, S.; Houlden, H. Neurodegeneration with brain iron accumulation. Handb. Clin. Neurol. 2018, 145, 157–166. [Google Scholar]
  223. Damulina, A.; Pirpamer, L.; Soellradl, M.; Sackl, M.; Tinauer, C.; Hofer, E.; Enzinger, C.; Gesierich, B.; Duering, M.; Ropele, S. Cross-sectional and Longitudinal Assessment of Brain Iron Level in Alzheimer Disease Using 3-T MRI. Radiology 2020, 296, 619–626. [Google Scholar] [CrossRef] [PubMed]
  224. Pchitskaya, E.; Popugaeva, E.; Bezprozvanny, I. Calcium signaling and molecular mechanisms underlying neurodegenerative diseases. Cell Calcium 2018, 70, 87–94. [Google Scholar] [CrossRef]
  225. Ghadery, C.; Pirpamer, L.; Hofer, E.; Langkammer, C.; Petrovic, K.; Loitfelder, M.; Schwingenschuh, P.; Seiler, S.; Duering, M.; Jouvent, E. R2 * mapping for brain iron: Associations with cognition in normal aging. Neurobiol. Aging 2015, 36, 925–932. [Google Scholar] [CrossRef] [PubMed]
  226. Qin, Y.; Zhu, W.; Zhan, C.; Zhao, L.; Wang, J.; Tian, Q.; Wang, W. Investigation on positive correlation of increased brain iron deposition with cognitive impairment in Alzheimer disease by using quantitative MR R2′ mapping. J. Huazhong Univ. Sci. Technol. Med. Sci. 2011, 31, 578. [Google Scholar] [CrossRef]
  227. Moon, Y.; Han, S.-H.; Moon, W.-J. Patterns of brain iron accumulation in vascular dementia and Alzheimer’s dementia using quantitative susceptibility mapping imaging. J. Alzheimer’s Dis. 2016, 51, 737–745. [Google Scholar] [CrossRef] [PubMed]
  228. Barbosa, J.H.O.; Santos, A.C.; Tumas, V.; Liu, M.; Zheng, W.; Haacke, E.M.; Salmon, C.E.G. Quantifying brain iron deposition in patients with Parkinson’s disease using quantitative susceptibility mapping, R2 and R2. Magn. Reson. Imaging 2015, 33, 559–565. [Google Scholar] [CrossRef]
  229. Cheng, Q.; Huang, J.; Liang, J.; Ma, M.; Zhao, Q.; Lei, X.; Shi, C.; Luo, L. Evaluation of abnormal iron distribution in specific regions in the brains of patients with Parkinson’s disease using quantitative susceptibility mapping and R2* mapping. Exp. Ther. Med. 2020, 19, 3778–3786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Wieler, M.; Gee, M.; Martin, W.W. Longitudinal midbrain changes in early Parkinson’s disease: Iron content estimated from R2 */MRI. Park. Relat. Disord. 2015, 21, 179–183. [Google Scholar] [CrossRef] [PubMed]
  231. Du, G.; Lewis, M.M.; Styner, M.; Shaffer, M.L.; Sen, S.; Yang, Q.X.; Huang, X. Combined R2 * and diffusion tensor imaging changes in the substantia nigra in Parkinson’s disease. Mov. Disord. 2011, 26, 1627–1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Khalil, M.; Langkammer, C.; Pichler, A.; Pinter, D.; Gattringer, T.; Bachmaier, G.; Ropele, S.; Fuchs, S.; Enzinger, C.; Fazekas, F. Dynamics of brain iron levels in multiple sclerosis: A longitudinal 3T MRI study. Neurology 2015, 84, 2396–2402. [Google Scholar] [CrossRef] [PubMed]
  233. Khalil, M.; Langkammer, C.; Ropele, S.; Petrovic, K.; Wallner-Blazek, M.; Loitfelder, M.; Jehna, M.; Bachmaier, G.; Schmidt, R.; Enzinger, C. Determinants of brain iron in multiple sclerosis: A quantitative 3T MRI study. Neurology 2011, 77, 1691–1697. [Google Scholar] [CrossRef] [PubMed]
  234. Walsh, A.J.; Blevins, G.; Lebel, R.M.; Seres, P.; Emery, D.J.; Wilman, A.H. Longitudinal MR imaging of iron in multiple sclerosis: An imaging marker of disease. Radiology 2014, 270, 186–196. [Google Scholar] [CrossRef]
  235. Paling, D.; Tozer, D.; Wheeler-Kingshott, C.; Kapoor, R.; Miller, D.H.; Golay, X. Reduced R2′ in multiple sclerosis normal appearing white matter and lesions may reflect decreased myelin and iron content. J. Neurol. Neurosurg. Psychiatry 2012, 83, 785–792. [Google Scholar] [CrossRef] [PubMed]
  236. Seif, M.; Curt, A.; Thompson, A.J.; Grabher, P.; Weiskopf, N.; Freund, P. Quantitative MRI of rostral spinal cord and brain regions is predictive of functional recovery in acute spinal cord injury. NeuroImage: Clin. 2018, 20, 556–563. [Google Scholar] [CrossRef] [PubMed]
  237. Blomster, L.V.; Cowin, G.J.; Kurniawan, N.D.; Ruitenberg, M.J. Detection of endogenous iron deposits in the injured mouse spinal cord through high-resolution ex vivo and in vivo MRI. NMR Biomed. 2013, 26, 141–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Haacke, E.M.; Mittal, S.; Wu, Z.; Neelavalli, J.; Cheng, Y.-C.N. Susceptibility-weighted imaging: Technical aspects and clinical applications, part 1. Am. J. Neuroradiol. 2009, 30, 19–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Mittal, S.; Wu, Z.; Neelavalli, J.; Haacke, E.M. Susceptibility-weighted imaging: Technical aspects and clinical applications, part 2. Am. J. Neuroradiol. 2009, 30, 232–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Halefoglu, A.M.; Yousem, D.M. Susceptibility weighted imaging: Clinical applications and future directions. World J. Radiol. 2018, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  241. Haacke, E.M.; Xu, Y.; Cheng, Y.C.N.; Reichenbach, J.R. Susceptibility weighted imaging (SWI). Magn. Reson. Med. 2004, 52, 612–618. [Google Scholar] [CrossRef]
  242. Yuste, R.; Majewska, A.; Holthoff, K. From form to function: Calcium compartmentalization in dendritic spines. Nat. Neurosci. 2000, 3, 653–659. [Google Scholar] [CrossRef] [PubMed]
  243. Burnashev, N.; Rozov, A. Presynaptic Ca2+ dynamics, Ca2+ buffers and synaptic efficacy. Cell Calcium 2005, 37, 489–495. [Google Scholar] [CrossRef] [PubMed]
  244. Hartmann, J.; Konnerth, A. Determinants of postsynaptic Ca2+ signaling in Purkinje neurons. Cell Calcium 2005, 37, 459–466. [Google Scholar] [CrossRef] [PubMed]
  245. Hidalgo, C.; Nunez, M.T. Calcium, iron and neuronal function. IUBMB Life 2007, 59, 280–285. [Google Scholar] [CrossRef] [PubMed]
  246. Hidalgo, C.; Carrasco, M.A.; Muñoz, P.; Núñez, M.T. A role for reactive oxygen/nitrogen species and iron on neuronal synaptic plasticity. Antioxid. Redox Signal. 2007, 9, 245–255. [Google Scholar] [CrossRef] [PubMed]
  247. Muñoz, P.; Humeres, A.; Elgueta, C.; Kirkwood, A.; Hidalgo, C.; Núñez, M.T. Iron mediates N-methyl-D-aspartate receptor-dependent stimulation of calcium-induced pathways and hippocampal synaptic plasticity. J. Biol. Chem. 2011, 286, 13382–13392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Hsu, C.Y.; Hogan, E.; Gadsden Sr, R.; Spicer, K.; Shi, M.; Cox, R. Vascular permeability in experimental spinal cord injury. J. Neurol. Sci. 1985, 70, 275–282. [Google Scholar] [CrossRef]
  249. Happel, R.D.; Smith, K.P.; Banik, L.N.; Powers, M.J.; Hogan, E.L.; Balentine, J.D. Ca2+—Accumulation in experimental spinal cord trauma. Brain Res. 1981, 211, 476–479. [Google Scholar] [CrossRef]
  250. Young, W.; Koreh, I. Potassium and calcium changes in injured spinal cords. Brain Res. 1986, 365, 42–53. [Google Scholar] [CrossRef]
  251. Mohammed, W.; Xunning, H.; Haibin, S.; Jingzhi, M. Clinical applications of susceptibility-weighted imaging in detecting and grading intracranial gliomas: A review. Cancer Imaging 2013, 13, 186–195. [Google Scholar] [CrossRef] [Green Version]
  252. Wu, Z.; Mittal, S.; Kish, K.; Yu, Y.; Hu, J.; Haacke, E.M. Identification of calcification with MRI using susceptibility-weighted imaging: A case study. J. Magn. Reson. Imaging 2009, 29, 177–182. [Google Scholar] [CrossRef] [Green Version]
  253. Robinson, R.J.; Bhuta, S. Susceptibility-Weighted Imaging of the Brain: Current Utility and Potential Applications. J. Neuroimaging 2011, 21, e189–e204. [Google Scholar] [CrossRef]
  254. Thomas, B.; Somasundaram, S.; Thamburaj, K.; Kesavadas, C.; Kumar Gupta, A.; Bodhey, N.K.; Raman Kapilamoorthy, T. Clinical applications of susceptibility weighted MR imaging of the brain—A pictorial review. Neuroradiology 2008, 50, 105–116. [Google Scholar] [CrossRef]
  255. Nair, J.R.; Van Hecke, W.; De Belder, F.; Venstermans, C.; van den Hauwe, L.; Van Goethem, J.; Parizel, P.M. High-Resolution Susceptibility-Weighted Imaging at 3 T With a 32-Channel Head Coil: Technique and Clinical Applications. Am. J. Roentgenol. 2010, 195, 1007–1014. [Google Scholar] [CrossRef]
  256. Benzel, E.C.; Lancon, J.; Kesterson, L.; Hadden, T. Cervical laminectomy and dentate ligament section for cervical spondylotic myelopathy. J. Spinal Disord. 1991, 4, 286–295. [Google Scholar] [CrossRef] [PubMed]
  257. McCaffery, M.; Beebe, A. The Numeric Pain Rating Scale Instructions Pain: Clinic Manual for Nursing Practice; 1989; Available online: http://nperesource.casn.ca/wp-content/uploads/2017/02/Numeric-Pain-Rating-Scale-Instructions.pdf (accessed on 24 August 2022).
  258. Vernon, H.; Mior, S. The Neck Disability Index: A study of reliability and validity. J. Manip. Physiol. Ther. 1991, 14, 409–415. [Google Scholar]
  259. Devlin, N.; Parkin, D.; Janssen, B. An introduction to EQ-5D instruments and their applications. In Methods for Analysing and Reporting EQ-5D Data; Springer: Cham, Switzerland, 2020; pp. 1–22. [Google Scholar]
  260. Nurjck, S. The pathogenesis of the spinal cord disorder associated with cervical spondylosis. Brain 1972, 95, 87–100. [Google Scholar] [CrossRef] [PubMed]
  261. Chiles, B.W., III; Leonard, M.A.; Choudhri, H.F.; Cooper, P.R. Cervical spondylotic myelopathy: Patterns of neurological deficit and recovery after anterior cervical decompression. Neurosurgery 1999, 44, 762–769. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The pathogenesis of DCM. A combination of dynamic and static stressors is purported to contribute to the development of DCM. Neural structure compression includes spinal canal compression, spine deformity, disc herniation, osteophyte formation, ossification of the posterior longitudinal ligaments (OPLL) and ossification of the ligamentum flavum (OFL). Dynamic stressors include biomechanical changes, invagination of the ligamentum flavum and microstructural mechanical spinal cord damage from cervical instability. Abbreviations: DCM, degenerative cervical myelopathy.
Figure 1. The pathogenesis of DCM. A combination of dynamic and static stressors is purported to contribute to the development of DCM. Neural structure compression includes spinal canal compression, spine deformity, disc herniation, osteophyte formation, ossification of the posterior longitudinal ligaments (OPLL) and ossification of the ligamentum flavum (OFL). Dynamic stressors include biomechanical changes, invagination of the ligamentum flavum and microstructural mechanical spinal cord damage from cervical instability. Abbreviations: DCM, degenerative cervical myelopathy.
Biomedicines 10 02621 g001
Figure 2. Where novel qMRI would fit into the current diagnostic work-up and treatment of degenerative cervical myelopathy. The dashed lines represent pathways currently under investigation. The current diagnostic work-up starts with a detailed history, physical examination, and application of scoring systems. Electrophysiology and imaging can rule out differentials and provide additional information to generate a diagnosis of DCM or cervical spinal cord compression without myelopathy. Surgery is recommended for moderate to severe myelopathy according to current guidelines. Patients with spinal cord compression and evidence of radiculopathy may be offered surgical or non-surgical treatment. Patients with spinal cord compression and no evidence of radiculopathy should undergo clinical monitoring. Surgery may be offered to patients utilising non-operative treatment upon worsening of condition. There is potential for qMRI to play a role in monitoring this progression and provide prognostic value to the outcome of DCM. Abbreviations: CMS, cervical myelopathy score; CT, computed tomography; DCM, Degenerative cervical myelopathy; DTI, Diffusion tensor imaging; EMG, electromyography; EMS, European myelopathy score; ENG, electroneurography; fMRI, functional MRI; MEPs, motor evoked potentials; mJOA, modified Japanese orthopaedic association score; MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; MT, magnetization transfer; NCS, nerve conduction studies; NDI, neck disability index; NPRS, numeric pain rating scale; qMRI, quantitative magnetic resonance imaging; qT1, quantitative T1; qT2, quantitative T2; SEPs, somatosensory evoked potentials; SWI, susceptibility weighted imaging.
Figure 2. Where novel qMRI would fit into the current diagnostic work-up and treatment of degenerative cervical myelopathy. The dashed lines represent pathways currently under investigation. The current diagnostic work-up starts with a detailed history, physical examination, and application of scoring systems. Electrophysiology and imaging can rule out differentials and provide additional information to generate a diagnosis of DCM or cervical spinal cord compression without myelopathy. Surgery is recommended for moderate to severe myelopathy according to current guidelines. Patients with spinal cord compression and evidence of radiculopathy may be offered surgical or non-surgical treatment. Patients with spinal cord compression and no evidence of radiculopathy should undergo clinical monitoring. Surgery may be offered to patients utilising non-operative treatment upon worsening of condition. There is potential for qMRI to play a role in monitoring this progression and provide prognostic value to the outcome of DCM. Abbreviations: CMS, cervical myelopathy score; CT, computed tomography; DCM, Degenerative cervical myelopathy; DTI, Diffusion tensor imaging; EMG, electromyography; EMS, European myelopathy score; ENG, electroneurography; fMRI, functional MRI; MEPs, motor evoked potentials; mJOA, modified Japanese orthopaedic association score; MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; MT, magnetization transfer; NCS, nerve conduction studies; NDI, neck disability index; NPRS, numeric pain rating scale; qMRI, quantitative magnetic resonance imaging; qT1, quantitative T1; qT2, quantitative T2; SEPs, somatosensory evoked potentials; SWI, susceptibility weighted imaging.
Biomedicines 10 02621 g002
Figure 3. Literature Search Flowchart (See Appendix BFigure A1. for database search strategy). Adapted from PRISMA Scoping Review protocol [121]. Abbreviations: MRI, magnetic resonance imaging.
Figure 3. Literature Search Flowchart (See Appendix BFigure A1. for database search strategy). Adapted from PRISMA Scoping Review protocol [121]. Abbreviations: MRI, magnetic resonance imaging.
Biomedicines 10 02621 g003
Figure 4. Preliminary data obtained from a healthy recruit using 3T MAGNETOM PRISMA MRI scanner with rFOV DTI ZOOMit sequence. From our standardisation work we have found rFOV to offer better visibility, better signal-to-noise, and less susceptibility and motion artifacts when compared to full field of view (fFOV) sequences. (a) axial Col-FA map of C4/5 cervical spinal-cord (red = left-right, blue = supra-inferior green = antero-posterior). (b) axial ADC map of C4/5 cervical spinal cord. Abbreviations: ADC, apparent diffusion coefficient; DTI, diffusion tensor imaging; Col-FA, colour fractional anisotropy; MRI, magnetic resonance imaging; rFOV, reduced field of view; ROI, region of interest.
Figure 4. Preliminary data obtained from a healthy recruit using 3T MAGNETOM PRISMA MRI scanner with rFOV DTI ZOOMit sequence. From our standardisation work we have found rFOV to offer better visibility, better signal-to-noise, and less susceptibility and motion artifacts when compared to full field of view (fFOV) sequences. (a) axial Col-FA map of C4/5 cervical spinal-cord (red = left-right, blue = supra-inferior green = antero-posterior). (b) axial ADC map of C4/5 cervical spinal cord. Abbreviations: ADC, apparent diffusion coefficient; DTI, diffusion tensor imaging; Col-FA, colour fractional anisotropy; MRI, magnetic resonance imaging; rFOV, reduced field of view; ROI, region of interest.
Biomedicines 10 02621 g004
Figure 5. Preliminary data obtained from a healthy recruit using 3T MAGNETOM PRISMA MRI scanner with R2* MRI. (a) axial R2* map of C2/3 cervical spinal-cord. (b) axial R2* map of C4/5 cervical spinal-cord. (c) axial R2* map of C2/3 cervical spinal-cord. Abbreviations: DCM, degenerative cervical myelopathy; MRI, magnetic resonance imaging; ROI, region of interest.
Figure 5. Preliminary data obtained from a healthy recruit using 3T MAGNETOM PRISMA MRI scanner with R2* MRI. (a) axial R2* map of C2/3 cervical spinal-cord. (b) axial R2* map of C4/5 cervical spinal-cord. (c) axial R2* map of C2/3 cervical spinal-cord. Abbreviations: DCM, degenerative cervical myelopathy; MRI, magnetic resonance imaging; ROI, region of interest.
Biomedicines 10 02621 g005aBiomedicines 10 02621 g005b
Table 1. Typical presenting symptoms and physical signs in DCM [2,37,38,39,40,41,42,43].
Table 1. Typical presenting symptoms and physical signs in DCM [2,37,38,39,40,41,42,43].
Presenting Symptoms Physical Signs
Neck
Pain and/or stiffness
Decreased cervical range of motion
The Lhermitte phenomenon [a]
Corticospinal tract distribution motor deficits
Upper Limb
Weakness
Pain
Paraesthesia
Upper motor neuron signs (hyper-reflexia, a positive Hoffman sign [b], a positive Trömner sign [c])
Sensory loss in a dermatomal pattern
Intrinsic hand muscle atrophy
Corticospinal tract distribution motor deficits
Lower Limb
Weakness
Pain
Paraesthesia
Loss of manual dexterity
Falls
Gait imbalance
Upper motor neuron signs (hyper-reflexia, a positive Babinski sign [d])
An unstable, broad-based gait
Sensory loss in a dermatomal pattern
Spasticity and clonus
Corticospinal tract distribution motor deficits
Urinary/defecatory
Frequency/urgency
Urge incontinence
Nil
Abbreviations: DCM, degenerative cervical myelopathy. [a] Lhermitte phenomenon: an electric shock-like sensation radiating from the neck down into the back that occurs upon flexion of the neck; [b] Positive Hoffman sign: flexion and adduction of the thumb and flexion of the index finger upon forceful flicking of the middle fingernail; [c] Positive Trömner sign: flexion of the thumb and index finger in response to flicking of the volar surface of the distal phalanx of the middle finger; [d] Positive Babinski sign: an upwards plantar response involving toe flexion after elicitation from the sole of the foot.
Table 2. Common classification systems used for DCM [17,47,48,49,50,51,52,53,54,55,56]. See Appendix A (Table A1, Table A2, Table A3, Table A4, Table A5, Table A6 and Table A7.) for full scoring systems.
Table 2. Common classification systems used for DCM [17,47,48,49,50,51,52,53,54,55,56]. See Appendix A (Table A1, Table A2, Table A3, Table A4, Table A5, Table A6 and Table A7.) for full scoring systems.
SystemDescriptionBenefitsLimitations
mJOA scale
0–18. A lower score indicates a more severe deficit.
Normal: 18
Mild myelopathy: 15–17
Mild myelopathy: 15–17
Moderate myelopathy: 12–14
Severe myelopathy: 0–11
Upper-extremity function (5)
Lower-extremity function (7)
Sensory function (3)
Bladder function (3)
Good for assessing outcomes (post-operative).
Specific to DCM
Responsive to change
Commonly used in research
Clinician administered
No economic factors taken into consideration.
Poor sensitivity
Ceiling effect: hard to detect minor improvements in patients with mild disease
Modest intra-rater and inter-rater reliability (the minimum detectable change is two points).
Four categories are not equally weighted.
Nurick scale
0–5. A higher grade indicates a more severe deficit.
Myelopathy (6 points)
See Table A5. for grade definitions
Good for evaluating economic situation in conjunction with gait function.
Specific to DCM
Commonly used in research
Consists of both impairment and disability components
Low sensitivity
Poor responsiveness with limited ability to detect change.
Less accurate for post-operative grading.
Cannot detect upper extremity dysfunction.
NDI
0–50. A higher grade indicates a more severe disability.
Neck disability (10 subsections)
0 = no disability5 = complete disability
Consists of: Pain intensity, personal care, lifting, reading, headaches, concentration, work, driving, sleeping, recreation
Fair interobserver reliability in patients that have cervical radiculopathy
Responsive to change
Incorporates various activities from daily living
Validity and reliability only evaluated in neck pain patients and cervical radiculopathy patients
Subjective; patient reported
Not specific to DCM
EMS
5–18. A lower score indicates a more severe deficit.
Normal: 17+
Grade 1: 13–16
Grade 2: 9–12
Grade 3: 5–8
Good at evaluating clinical state and level of severity.
Better sensitivity towards functional deficits (as it assesses coordination and proprioception)
Not commonly used in research
CMS
Upper/lower extremities are analysed separately 0–5 each.
A higher grade indicates a more severe deficit.
Good for evaluating upper/lower extremity function as they are elicited separately.
Good at evaluating clinical state and level of severity.
No economic factors taken into consideration.
NPRS
0–10. A higher score indicates a more severe disability
Simplicity and reproducibility
Sensitive to small changes
Valid
Not specific to DCM
Subjective
Suffers from the ceiling effect
EQ-5D
A standardised measure of health status looking into mobility, self-care, activities of daily living, pain/discomfort, anxiety/depression.
Not measured on a numbered scale
Ease of completion
Sensitive to change
Useful for looking into health economic evaluations
Emotions and mood are limited to anxiety and depression
Quite global in nature
Overlooks some dimensions of quality of life (spiritual, social)
Does not include cognition
Not specific to DCM
Additional scales that provide useful information in the context of DCM include the Myelopathy Disability Index, QuickDASH (assesses arm, shoulder and hand disability), the 30-Metre-Walk test, the Berg Balance Scale, GAITRite (a temporospatial gait analysis) and the Graded Redefined Assessment of Strength Sensibility and Prehension Myelopathy (GRASSP-M).
Abbreviations: CMS, Cervical Myelopathy Scale; DCM, degenerative cervical myelopathy; EMS, European Myelopathy Scale; mJOA, modified Japanese Orthopaedic Association; NDI, Neck Disability Index; NPRS, Numeric Pain Rating Scale.
Table 3. Quantitative MRI sequences applicable in the context of myelopathy and their corresponding derived metrics [70,103,104,105,106,107,110,111,112,113,114,115,116,117,118,119,120].
Table 3. Quantitative MRI sequences applicable in the context of myelopathy and their corresponding derived metrics [70,103,104,105,106,107,110,111,112,113,114,115,116,117,118,119,120].
SequenceFunctionQuantitative Metrics
Quantitative T1/T2 MappingCalculates the T1/T2 time of certain tissues and displays them on a parametric map. Reveals information about microstructural changes related to water, lipid, protein and iron content of tissues.T1/T2 relaxation time
DWIDTIEstimates the integrity of tissue microstructure through the modelling of water diffusion within the tissue.FA [f], ADC, MD [g]
DTTTracks nerve fibres based on their FA values and can be elicited when fibres become interrupted, distorted or disorientated depending on the severity of spinal compression.Volume and number of fibres
DBSIQuantifies axonal injury, inflammation and demyelination in DCMAxonal injury, inflammation, demyelination.
fMRI (BOLD)Measures neuronal activity through associated changes detected in blood flowFC, VOA
MTProvides information on the spinal cord structural integrity and derive information regarding myelination statusMTR
MRSSensitive to metabolic changes that occur in pathology, reflecting important underlying biological mechanismsMetabolite concentrations
T2*-weighted imagingQuantifies observable or effective T2 and is utilised to detect deoxyhaemoglobin, hemosiderin or methemoglobin in tissues and lesions.R2* (=1/T2*)
SWI/QSMSensitive to compounds that distort the magnetic field and alter phase of tissue and is therefore commonly used to detect blood products/haemorrhage and calciumTissue susceptibility
Abbreviations: ADC, apparent diffusion coefficient; BOLD, blood oxygen level dependent; DBSI, diffusion basis spectrum imaging; DCM, degenerative cervical myelopathy; DTI, diffusion tensor imaging; DTT, diffusion tensor tractography; DWI, diffusion weighted imaging; FA, fractional anisotropy; FC, functional connectivity; fMRI, functional magnetic resonance imaging; MD, mean diffusivity; MRS, magnetic resonance spectroscopy; MT, magnetisation transfer; MTR, magnetisation transfer ratio; QSM, quantitative susceptibility mapping; SWI, susceptibility weighted imaging; VOA, volume of activation. [f] Fractional anisotropy (FA): Water molecules diffuse differently along tissues depending on its type, integrity, architecture, and presence of barriers. Fractional anisotropy is a value between 0 and 1 which indicates the degree to which diffusion of water is limited to one axis; [g] Apparent diffusion coefficient (ADC)/mean diffusivity (MD): measures of the average magnitude of water diffusion within a tissue.
Table 4. Summary of qMRI techniques utilised in the 68 included articles of this study (N.B. some studies investigated a multiplicity of qMRI techniques). Refer to Appendix C (Table A8) for included articles [60,104,105,108,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185].
Table 4. Summary of qMRI techniques utilised in the 68 included articles of this study (N.B. some studies investigated a multiplicity of qMRI techniques). Refer to Appendix C (Table A8) for included articles [60,104,105,108,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185].
qMRI Technique UtilisedNumber of StudiesOverall Findings from the Included Literature
Quantitative T12
Higher T1 values in spinal cord of moderate-severe DCM
Inconclusive/variables results about mild cervical cord stenosis and mild DCM
Studies limited by low sample size and cross-sectional design
Quantitative T20Nil
DTI43
Strong correlation of FA and mJOA
FA as a significant prognostic indicator
Need for more longitudinal large sample-size studies with longer time-horizons
DTI ratios as a better assessment metric than absolute DTI value.
fMRI (BOLD)15
Successful in differentiating DCM patients from healthy controls
Positive correlation of FC and VOA with various connections of the brain with post-surgical recovery
Notable correlation with visual cortices
Further external validation studies necessary
A need for prospective studies over 6 months to be conducted
MRS6
Cho/Naa ratio presents the best correlation with DCM severity.
Limitations with acquisition reliability
No new literature conducted in past 6 years
All cross-sectional and of low sample size
Further longitudinal and prognostic studies necessary
MT4
MTR negatively correlates with DCM severity
Potential confounding of data with MTR being additionally associated with age related demyelination
Low sample sizes
Overall limited research in this area, further longitudinal prospective studies required
R2* or 1/T2*0Nil
SWI0Nil
Abbreviations: BOLD, blood oxygen level dependent; Cho, choline; DCM, degenerative cervical myelopathy; DTI, diffusion tensor imaging; FA, fractional anisotropy; FC, functional connectivity; fMRI, functional magnetic resonance imaging; mJOA, modified Japanese Orthopaedic Association scale; MRS, magnetic resonance spectroscopy; MT, magnetisation transfer; MTR, magnetisation transfer ratio; NAA, n-acetylaspartate; qMRI, quantitative magnetic resonance imaging; SWI, susceptibility weighted imaging; VOA, volume of activation.
Table 5. Preliminary data obtained from our initial incomplete study, showcasing FA and ADC ROI scores of a healthy recruit. Data obtained through a standardisation protocol of DTI using 3T MAGNETOM PRISMA MRI scanner with a rFOV ZOOMit sequence with 4 averages (dynamic excitation for selective centrally excited field of view). We expect FA to be lower in recruits with DCM.
Table 5. Preliminary data obtained from our initial incomplete study, showcasing FA and ADC ROI scores of a healthy recruit. Data obtained through a standardisation protocol of DTI using 3T MAGNETOM PRISMA MRI scanner with a rFOV ZOOMit sequence with 4 averages (dynamic excitation for selective centrally excited field of view). We expect FA to be lower in recruits with DCM.
MeasurementsMin/Max (×10−3)Mean (×10−3)Standard Deviation (×10−3)Area (cm2)
FA219/1000629.16201.720.35
ADC186/1222752.89238.790.35
Abbreviations: ADC, apparent diffusion coefficient; DCM, degenerative cervical myelopathy; DTI, diffusion tensor imaging; FA, fractional anisotropy; MRI, magnetic resonance imaging; rFOV, reduced field of view; ROI, region of interest.
Table 6. Preliminary data obtained from our initial incomplete study, showcasing R2* region of interest (ROI) scores of a healthy recruit. Data obtained through a standardisation protocol of R2* using 3T MAGNETOM PRISMA MRI scanner. The values obtained from our R2* is equivalent to other soft tissues in the absence of pathological processes leading to iron deposition (based on R2* studies done in the brain and liver) which tends around 30. As such, we would expect higher R2* values in recruits with DCM.
Table 6. Preliminary data obtained from our initial incomplete study, showcasing R2* region of interest (ROI) scores of a healthy recruit. Data obtained through a standardisation protocol of R2* using 3T MAGNETOM PRISMA MRI scanner. The values obtained from our R2* is equivalent to other soft tissues in the absence of pathological processes leading to iron deposition (based on R2* studies done in the brain and liver) which tends around 30. As such, we would expect higher R2* values in recruits with DCM.
Cervical LevelMin/Max (1/s)Mean (1/s)Standard DeviationArea (cm2)
C2/318.00/30.0023.413.030.60
C3/412.00/30.0023.173.260.59
C4/518.00/44.0031.404.200.43
Abbreviations: DCM, degenerative cervical myelopathy; MRI, magnetic resonance imaging; ROI, region of interest.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

He, B.; Sheldrick, K.; Das, A.; Diwan, A. Clinical and Research MRI Techniques for Assessing Spinal Cord Integrity in Degenerative Cervical Myelopathy—A Scoping Review. Biomedicines 2022, 10, 2621. https://doi.org/10.3390/biomedicines10102621

AMA Style

He B, Sheldrick K, Das A, Diwan A. Clinical and Research MRI Techniques for Assessing Spinal Cord Integrity in Degenerative Cervical Myelopathy—A Scoping Review. Biomedicines. 2022; 10(10):2621. https://doi.org/10.3390/biomedicines10102621

Chicago/Turabian Style

He, Brandon, Kyle Sheldrick, Abhirup Das, and Ashish Diwan. 2022. "Clinical and Research MRI Techniques for Assessing Spinal Cord Integrity in Degenerative Cervical Myelopathy—A Scoping Review" Biomedicines 10, no. 10: 2621. https://doi.org/10.3390/biomedicines10102621

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop